171
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The Major Cellular Sterol Regulatory Pathway Is Required for Andes Virus Infection

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The Bunyaviridae comprise a large family of RNA viruses with worldwide distribution and includes the pathogenic New World hantavirus, Andes virus (ANDV). Host factors needed for hantavirus entry remain largely enigmatic and therapeutics are unavailable. To identify cellular requirements for ANDV infection, we performed two parallel genetic screens. Analysis of a large library of insertionally mutagenized human haploid cells and a siRNA genomic screen converged on components (SREBP-2, SCAP, S1P and S2P) of the sterol regulatory pathway as critically important for infection by ANDV. The significance of this pathway was confirmed using functionally deficient cells, TALEN-mediated gene disruption, RNA interference and pharmacologic inhibition. Disruption of sterol regulatory complex function impaired ANDV internalization without affecting virus binding. Pharmacologic manipulation of cholesterol levels demonstrated that ANDV entry is sensitive to changes in cellular cholesterol and raises the possibility that clinically approved regulators of sterol synthesis may prove useful for combating ANDV infection.

          Author Summary

          As obligate, intracellular parasites viruses are dependent upon the host cell for numerous factors and processes. However, for many important viruses few of the required host factors have been identified. Hantaviruses are rodent-borne viruses that are associated with severe human disease. Transmission to humans occurs sporadically with a recent notable example in Yosemite National park. In the present study, we utilized two independent genetic strategies to discover cellular factors needed for replication of the highly pathogenic hantavirus Andes virus. We found that four genes, encoding components of a complex involved in regulation of cholesterol synthesis and uptake, were critical for Andes virus infection. Drugs that inhibit an enzyme in this complex or that reduce cellular cholesterol levels effectively blocked Andes virus infection, suggesting new ways for combating this pathogenic virus.

          Related collections

          Most cited references47

          • Record: found
          • Abstract: found
          • Article: not found

          Virus entry by endocytosis.

          Although viruses are simple in structure and composition, their interactions with host cells are complex. Merely to gain entry, animal viruses make use of a repertoire of cellular processes that involve hundreds of cellular proteins. Although some viruses have the capacity to penetrate into the cytosol directly through the plasma membrane, most depend on endocytic uptake, vesicular transport through the cytoplasm, and delivery to endosomes and other intracellular organelles. The internalization may involve clathrin-mediated endocytosis (CME), macropinocytosis, caveolar/lipid raft-mediated endocytosis, or a variety of other still poorly characterized mechanisms. This review focuses on the cell biology of virus entry and the different strategies and endocytic mechanisms used by animal viruses.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Interferon-inducible cholesterol-25-hydroxylase broadly inhibits viral entry by production of 25-hydroxycholesterol.

            Interferons (IFN) are essential antiviral cytokines that establish the cellular antiviral state through upregulation of hundreds of interferon-stimulated genes (ISGs), most of which have uncharacterized functions and mechanisms. We identified cholesterol-25-hydroxylase (CH25H) as a broadly antiviral ISG. CH25H converts cholesterol to a soluble antiviral factor, 25-hydroxycholesterol (25HC). 25HC treatment in cultured cells broadly inhibited growth of enveloped viruses including VSV, HSV, HIV, and MHV68 and acutely pathogenic EBOV, RVFV, RSSEV, and Nipah viruses under BSL4 conditions. It suppressed viral growth by blocking membrane fusion between virus and cell. In animal models, Ch25h-deficient mice were more susceptible to MHV68 lytic infection. Moreover, administration of 25HC in humanized mice suppressed HIV replication and reversed T cell depletion. Thus, our studies demonstrate a unique mechanism by which IFN achieves its antiviral state through the production of a natural oxysterol to inhibit viral entry and implicate membrane-modifying oxysterols as potential antiviral therapeutics. Copyright © 2013 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Extraction of cholesterol with methyl-beta-cyclodextrin perturbs formation of clathrin-coated endocytic vesicles.

              The importance of cholesterol for endocytosis has been investigated in HEp-2 and other cell lines by using methyl-beta-cyclodextrin (MbetaCD) to selectively extract cholesterol from the plasma membrane. MbetaCD treatment strongly inhibited endocytosis of transferrin and EGF, whereas endocytosis of ricin was less affected. The inhibition of transferrin endocytosis was completely reversible. On removal of MbetaCD it was restored by continued incubation of the cells even in serum-free medium. The recovery in serum-free medium was inhibited by addition of lovastatin, which prevents cholesterol synthesis, but endocytosis recovered when a water-soluble form of cholesterol was added together with lovastatin. Electron microscopical studies of MbetaCD-treated HEp-2 cells revealed that typical invaginated caveolae were no longer present. Moreover, the invagination of clathrin-coated pits was strongly inhibited, resulting in accumulation of shallow coated pits. Quantitative immunogold labeling showed that transferrin receptors were concentrated in coated pits to the same degree (approximately sevenfold) after MbetaCD treatment as in control cells. Our results therefore indicate that although clathrin-independent (and caveolae-independent) endocytosis still operates after removal of cholesterol, cholesterol is essential for the formation of clathrin-coated endocytic vesicles.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Pathog
                PLoS Pathog
                plos
                plospath
                PLoS Pathogens
                Public Library of Science (San Francisco, USA )
                1553-7366
                1553-7374
                February 2014
                6 February 2014
                : 10
                : 2
                : e1003911
                Affiliations
                [1 ]Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
                [2 ]Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
                Mount Sinai School of Medicine, United States of America
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: JP MJD EAB AMR CAD CBW SC FDB RWD PB KB. Performed the experiments: JP MJD EAB AMR CAD CBW NM FM FHL KB. Analyzed the data: JP MJD EAB AMR CAD CBW NM FM FHL FDB SC RWD PB KB. Contributed reagents/materials/analysis tools: NM FM FDB. Wrote the paper: JP MJD EAB AMR SC RWD KB PB.

                Article
                PPATHOGENS-D-13-01777
                10.1371/journal.ppat.1003911
                3916400
                24516383
                07477da7-0e8a-4302-9f16-ece4d5eb5cb4
                Copyright @ 2014

                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.

                History
                : 8 July 2013
                : 18 December 2013
                Page count
                Pages: 13
                Funding
                This work was supported by grants from the National Institutes of Health R01AI081913 (PB), R01AI074951 (SC) U54AI057168 (RWD & SC) and R01AI095500 (SC). Trainees were supported by NIH awards T32AI055400 (JP and KB), T32AI007324 (EAB), T32GM007229 (MJD), and T32AI07632 (CBW). This work was also supported by R01 AI052845 (to FDB) and F32 AI106333 (to KB). SC is a recipient of the Burroughs Wellcome Investigators in the Pathogenesis of Infectious Disease Award. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology
                Genetics
                Genetic Screens
                Microbiology
                Pathogenesis
                Virology
                Molecular Cell Biology
                Membranes and Sorting
                Medicine
                Infectious Diseases
                Viral Diseases
                Hantavirus Pulmonary Syndrome

                Infectious disease & Microbiology
                Infectious disease & Microbiology

                Comments

                Comment on this article