10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      DDX17 Specifically, and Independently of DDX5, Controls Use of the HIV A4/5 Splice Acceptor Cluster and Is Essential for Efficient Replication of HIV

      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

          HIV splicing involves five splice donor and eight splice acceptor sequences which, together with cryptic splice sites, generate over 100 mRNA species. Ninety percent of both partially spliced and fully spliced transcripts utilize the intrinsically weak A4/A5 3′ splice site cluster. We show that DDX17, but not its close paralog DDX5, specifically controls the usage of this splice acceptor group. In its absence, production of the viral envelope protein and other regulatory and accessory proteins is grossly reduced, while Vif, which uses the A1 splice acceptor, is unaffected. This is associated with a profound decrease in viral export from the cell. Loss of Vpu expression causing upregulation of cellular Tetherin compounds the phenotype. DDX17 utilizes distinct RNA binding motifs for its role in efficient HIV replication, and we identify RNA binding motifs essential for its role, while the Walker A, Walker B (DEAD), Q motif and the glycine doublet motif are all dispensable. We show that DDX17 interacts with SRSF1/SF2 and the heterodimeric auxiliary factor U2AF65/35, which are essential splicing factors in the generation of Rev and Env/Vpu transcripts.

          Graphical Abstract

          Highlights

          • Regulation of HIV splicing is significantly dependent on host cellular factors.

          • DDX17 interacts with splicing factors and is essential for efficient HIV-1 splicing.

          • DDX17 utilizes distinct RNA/ATPase motifs and acts independently of DDX5.

          • DDX17 C-terminal domain is critical for the role of DDX17 in HIV-1 replication.

          Related collections

          Most cited references55

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

          Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu.

          Human cells possess an antiviral activity that inhibits the release of retrovirus particles, and other enveloped virus particles, and is antagonized by the HIV-1 accessory protein, Vpu. This antiviral activity can be constitutively expressed or induced by interferon-alpha, and it consists of protein-based tethers, which we term 'tetherins', that cause retention of fully formed virions on infected cell surfaces. Using deductive constraints and gene expression analyses, we identify CD317 (also called BST2 or HM1.24), a membrane protein of previously unknown function, as a tetherin. Specifically, CD317 expression correlated with, and induced, a requirement for Vpu during HIV-1 and murine leukaemia virus particle release. Furthermore, in cells where HIV-1 virion release requires Vpu expression, depletion of CD317 abolished this requirement. CD317 caused retention of virions on cell surfaces and, after endocytosis, in CD317-positive compartments. Vpu co-localized with CD317 and inhibited these effects. Inhibition of Vpu function and consequent mobilization of tetherin's antiviral activity is a potential therapeutic strategy in HIV/AIDS.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The interferon-induced protein BST-2 restricts HIV-1 release and is downregulated from the cell surface by the viral Vpu protein.

            The HIV-1 accessory protein Vpu counteracts a host factor that restricts virion release from infected cells. Here we show that the interferon-induced cellular protein BST-2/HM1.24/CD317 is such a factor. BST-2 is downregulated from the cell surface by Vpu, and BST-2 is specifically expressed in cells that support the vpu phenotype. Exogenous expression of BST-2 inhibits HIV-1 virion release, while suppression of BST-2 relieves the requirement for Vpu. Downregulation of BST-2 requires both the transmembrane/ion channel domain and conserved serines in the cytoplasmic domain of Vpu. Endogenous BST-2 colocalizes with the HIV-1 structural protein Gag in endosomes and at the plasma membrane, suggesting that BST-2 traps virions within and on infected cells. The unusual structure of BST-2, which includes a transmembrane domain and a lumenal GPI anchor, may allow it to retain nascent enveloped virions on cellular membranes, providing a mechanism of viral restriction counteracted by a specific viral accessory protein.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The DEAD-box protein family of RNA helicases.

              RNA helicases of the DEAD-box protein family have been shown to participate in every aspect of RNA metabolism. They are present in most organisms where they work as RNA helicases or RNPases. The properties of these enzymes in vivo remains poorly described, however some were extensively characterized in vitro, and the solved crystal structures of a few are now available. Taken together, this information gives insight into the regulation of ATP and RNA binding as well as in the ATPase and helicase activities. This review will focus on the description of the molecular characteristics of members of the DEAD-box protein family and on the enzymatic activities they possess.
                Bookmark

                Author and article information

                Contributors
                Journal
                J Mol Biol
                J. Mol. Biol
                Journal of Molecular Biology
                Elsevier
                0022-2836
                1089-8638
                14 September 2018
                14 September 2018
                : 430
                : 18Part B
                : 3111-3128
                Affiliations
                [1 ]Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
                [2 ]Department of Microbiology and Immunology, National University of Singapore, Singapore 117545
                [3 ]Department of Medicine, National University of Singapore, Singapore 119228
                Author notes
                [* ]Corresponding author. Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK. amll1@ 123456medschl.cam.ac.uk
                [†]

                Current Address: Specialist Virology Centre, Department of Microbiology, Norfolk and Norwich University Hospitals, Norwich, UK.

                [‡]

                Current address: Division of Virology, Department of Medicine, St Mary’s campus, Imperial College London, Norfolk Place, W2 1PG, London, UK.

                Article
                S0022-2836(18)30723-X
                10.1016/j.jmb.2018.06.052
                6119765
                30131116
                0e9ed855-0b30-4928-80e2-78a159ee5666
                © 2018 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 3 January 2018
                : 26 June 2018
                : 27 June 2018
                Categories
                Article

                Molecular biology
                ddx17/hiv-1,splicing factors,a4/a5 3′ splice site cluster,eses, exonic splicing enhancers,dapi, 4′,6-diamidino-2-phenlindole

                Comments

                Comment on this article