18
views
0
recommends
+1 Recommend
0 collections
    2
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Noroviruses Co-opt the Function of Host Proteins VAPA and VAPB for Replication via a Phenylalanine–Phenylalanine-Acidic-Tract-Motif Mimic in Nonstructural Viral Protein NS1/2

      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 Norovirus genus contains important human pathogens, but the role of host pathways in norovirus replication is largely unknown. Murine noroviruses provide the opportunity to study norovirus replication in cell culture and in small animals. The human norovirus nonstructural protein NS1/2 interacts with the host protein VAMP-associated protein A (VAPA), but the significance of the NS1/2-VAPA interaction is unexplored. Here we report decreased murine norovirus replication in VAPA- and VAPB-deficient cells. We characterized the role of VAPA in detail. VAPA was required for the efficiency of a step(s) in the viral replication cycle after entry of viral RNA into the cytoplasm but before the synthesis of viral minus-sense RNA. The interaction of VAPA with viral NS1/2 proteins is conserved between murine and human noroviruses. Murine norovirus NS1/2 directly bound the major sperm protein (MSP) domain of VAPA through its NS1 domain. Mutations within NS1 that disrupted interaction with VAPA inhibited viral replication. Structural analysis revealed that the viral NS1 domain contains a mimic of the phenylalanine–phenylalanine-acidic-tract (FFAT) motif that enables host proteins to bind to the VAPA MSP domain. The NS1/2-FFAT mimic region interacted with the VAPA-MSP domain in a manner similar to that seen with bona fide host FFAT motifs. Amino acids in the FFAT mimic region of the NS1 domain that are important for viral replication are highly conserved across murine norovirus strains. Thus, VAPA interaction with a norovirus protein that functionally mimics host FFAT motifs is important for murine norovirus replication.

          IMPORTANCE

          Human noroviruses are a leading cause of gastroenteritis worldwide, but host factors involved in norovirus replication are incompletely understood. Murine noroviruses have been studied to define mechanisms of norovirus replication. Here we defined the importance of the interaction between the hitherto poorly studied NS1/2 norovirus protein and the VAPA host protein. The NS1/2-VAPA interaction is conserved between murine and human noroviruses and was important for early steps in murine norovirus replication. Using structure-function analysis, we found that NS1/2 contains a short sequence that molecularly mimics the FFAT motif that is found in multiple host proteins that bind VAPA. This represents to our knowledge the first example of functionally important mimicry of a host FFAT motif by a microbial protein.

          Related collections

          Most cited references59

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          NMRFAM-SPARKY: enhanced software for biomolecular NMR spectroscopy

          Summary: SPARKY (Goddard and Kneller, SPARKY 3) remains the most popular software program for NMR data analysis, despite the fact that development of the package by its originators ceased in 2001. We have taken over the development of this package and describe NMRFAM-SPARKY, which implements new functions reflecting advances in the biomolecular NMR field. NMRFAM-SPARKY has been repackaged with current versions of Python and Tcl/Tk, which support new tools for NMR peak simulation and graphical assignment determination. These tools, along with chemical shift predictions from the PACSY database, greatly accelerate protein side chain assignments. NMRFAM-SPARKY supports automated data format interconversion for interfacing with a variety of web servers including, PECAN , PINE, TALOS-N, CS-Rosetta, SHIFTX2 and PONDEROSA-C/S. Availability and implementation: The software package, along with binary and source codes, if desired, can be downloaded freely from http://pine.nmrfam.wisc.edu/download_packages.html. Instruction manuals and video tutorials can be found at http://www.nmrfam.wisc.edu/nmrfam-sparky-distribution.htm. Contact: whlee@nmrfam.wisc.edu or markley@nmrfam.wisc.edu Supplementary information: Supplementary data are available at Bioinformatics online.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A four-step cycle driven by PI(4)P hydrolysis directs sterol/PI(4)P exchange by the ER-Golgi tether OSBP.

            Several proteins at endoplasmic reticulum (ER)-Golgi membrane contact sites contain a PH domain that interacts with the Golgi phosphoinositide PI(4)P, a FFAT motif that interacts with the ER protein VAP-A, and a lipid transfer domain. This architecture suggests the ability to both tether organelles and transport lipids between them. We show that in oxysterol binding protein (OSBP) these two activities are coupled by a four-step cycle. Membrane tethering by the PH domain and the FFAT motif enables sterol transfer by the lipid transfer domain (ORD), followed by back transfer of PI(4)P by the ORD. Finally, PI(4)P is hydrolyzed in cis by the ER protein Sac1. The energy provided by PI(4)P hydrolysis drives sterol transfer and allows negative feedback when PI(4)P becomes limiting. Other lipid transfer proteins are tethered by the same mechanism. Thus, OSBP-mediated back transfer of PI(4)P might coordinate the transfer of other lipid species at the ER-Golgi interface. Copyright © 2013 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Cholesterol sensor ORP1L contacts the ER protein VAP to control Rab7–RILP–p150Glued and late endosome positioning

              Late endosomes (LEs) have characteristic intracellular distributions determined by their interactions with various motor proteins. Motor proteins associated to the dynactin subunit p150Glued bind to LEs via the Rab7 effector Rab7-interacting lysosomal protein (RILP) in association with the oxysterol-binding protein ORP1L. We found that cholesterol levels in LEs are sensed by ORP1L and are lower in peripheral vesicles. Under low cholesterol conditions, ORP1L conformation induces the formation of endoplasmic reticulum (ER)–LE membrane contact sites. At these sites, the ER protein VAP (VAMP [vesicle-associated membrane protein]-associated ER protein) can interact in trans with the Rab7–RILP complex to remove p150Glued and associated motors. LEs then move to the microtubule plus end. Under high cholesterol conditions, as in Niemann-Pick type C disease, this process is prevented, and LEs accumulate at the microtubule minus end as the result of dynein motor activity. These data explain how the ER and cholesterol control the association of LEs with motor proteins and their positioning in cells.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mBio
                MBio
                mbio
                mbio
                mBio
                mBio
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2150-7511
                11 July 2017
                Jul-Aug 2017
                : 8
                : 4
                : e00668-17
                Affiliations
                [a ]Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
                [b ]Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
                [c ]Department of Pathology, Division of Virology, University of Cambridge, Cambridge, United Kingdom
                University of Pittsburgh School of Medicine
                Author notes
                Address correspondence to Herbert W. Virgin, virgin@ 123456wustl.edu .
                [*]

                Present address: Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA.

                This article is a direct contribution from a Fellow of the American Academy of Microbiology. External solicited reviewers: Ralph Baric, University of North Carolina at Chapel Hill; Daniele Fabris, The RNA Institute, State University of New York at Albany.

                Article
                mBio00668-17
                10.1128/mBio.00668-17
                5513711
                28698274
                e48d48b2-b3a4-4b90-9bfe-b88165ca8a43
                Copyright © 2017 McCune et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 29 April 2017
                : 4 May 2017
                Page count
                supplementary-material: 5, Figures: 7, Tables: 0, Equations: 1, References: 68, Pages: 17, Words: 10972
                Funding
                Funded by: HHS | NIH | National Cancer Institute (NCI) https://doi.org/10.13039/100000054
                Award ID: P30CA091842
                Award Recipient : Herbert W. Virgin
                Funded by: HHS | NIH | National Cancer Institute (NCI) https://doi.org/10.13039/100000054
                Award ID: F31CA177194-01
                Award Recipient : Broc T. McCune
                Funded by: HHS | National Institutes of Health (NIH) https://doi.org/10.13039/100000002
                Award ID: 5T32A100716334
                Award Recipient : Timothy J. Nice
                Funded by: HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID) https://doi.org/10.13039/100000060
                Award ID: U19 AI 109725
                Award Recipient : Herbert W. Virgin
                Funded by: American Cancer Society (ACS) https://doi.org/10.13039/100000048
                Award ID: Postdoc Fellowship
                Award Recipient : Timothy J. Nice
                Funded by: HHS | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) https://doi.org/10.13039/100000069
                Award ID: P30AR048335
                Award Recipient : Herbert W. Virgin
                Funded by: Cancer Research Institute (CRI) https://doi.org/10.13039/100000884
                Award ID: Postdoc Fellowship
                Award Recipient : Timothy J. Nice
                Funded by: Wellcome https://doi.org/10.13039/100010269
                Award ID: 097997/Z/11/Z
                Award Recipient : Ian G. Goodfellow
                Funded by: RCUK | Biotechnology and Biological Sciences Research Council (BBSRC) https://doi.org/10.13039/501100000268
                Award ID: BB/N001176/1
                Award Recipient : Ian G. Goodfellow
                Funded by: Winston Churchill Foundation of the United States (Winston Churchill Foundation) https://doi.org/10.13039/100008514
                Award ID: Churchill Scholarship
                Award Recipient : James B. Eaglesham
                Categories
                Research Article
                Custom metadata
                July/August 2017

                Life sciences
                noroviruses,plus-strand rna virus,protein structure-function,reverse genetic analysis,viral replication,virus-host interactions

                Comments

                Comment on this article