9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Interferon-Stimulated Gene (ISG)-Expression Screening Reveals the Specific Antibunyaviral Activity of ISG20

      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

          Bunyaviruses pose a significant threat to human health, prosperity, and food security. In response to viral infections, interferons (IFNs) upregulate the expression of hundreds of interferon-stimulated genes (ISGs), whose cumulative action can potently inhibit the replication of bunyaviruses. We used a flow cytometry-based method to screen the ability of ∼500 unique ISGs from humans and rhesus macaques to inhibit the replication of Bunyamwera orthobunyavirus (BUNV), the prototype of both the Peribunyaviridae family and the Bunyavirales order. Candidates possessing antibunyaviral activity were further examined using a panel of divergent bunyaviruses. Interestingly, one candidate, ISG20, exhibited potent antibunyaviral activity against most viruses examined from the Peribunyaviridae, Hantaviridae, and Nairoviridae families, whereas phleboviruses ( Phenuiviridae) largely escaped inhibition. Similar to the case against other viruses known to be targeted by ISG20, the antibunyaviral activity of ISG20 is dependent upon its functional RNase activity. Through use of an infectious virus-like particle (VLP) assay (based on the BUNV minigenome system), we confirmed that gene expression from all 3 viral segments is strongly inhibited by ISG20. Using in vitro evolution, we generated a substantially ISG20-resistant BUNV and mapped the determinants of ISG20 sensitivity/resistance. Taking all the data together, we report that ISG20 is a broad and potent antibunyaviral factor but that some bunyaviruses are remarkably ISG20 resistant. Thus, ISG20 sensitivity/resistance may influence the pathogenesis of bunyaviruses, many of which are emerging viruses of clinical or veterinary significance.

          IMPORTANCE There are hundreds of bunyaviruses, many of which cause life-threatening acute diseases in humans and livestock. The interferon (IFN) system is a key component of innate immunity, and type I IFNs limit bunyaviral propagation both in vitro and in vivo. Type I IFN signaling results in the upregulation of hundreds of IFN-stimulated genes (ISGs), whose concerted action generates an “antiviral state.” Although IFNs are critical in limiting bunyaviral replication and pathogenesis, much is still unknown about which ISGs inhibit bunyaviruses. Using ISG-expression screening, we examined the ability of ∼500 unique ISGs to inhibit Bunyamwera orthobunyavirus (BUNV), the prototypical bunyavirus. Using this approach, we identified ISG20, an interferon-stimulated exonuclease, as a potent inhibitor of BUNV. Interestingly, ISG20 possesses highly selective antibunyaviral activity, with multiple bunyaviruses being potently inhibited while some largely escape inhibition. We speculate that the ability of some bunyaviruses to escape ISG20 may influence their pathogenesis.

          Related collections

          Most cited references52

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

          IFIT1 is an antiviral protein that recognizes 5'-triphosphate RNA.

          Antiviral innate immunity relies on the recognition of microbial structures. One such structure is viral RNA that carries a triphosphate group on its 5' terminus (PPP-RNA). By an affinity proteomics approach with PPP-RNA as the 'bait', we found that the antiviral protein IFIT1 (interferon-induced protein with tetratricopeptide repeats 1) mediated binding of a larger protein complex containing other IFIT family members. IFIT1 bound PPP-RNA with nanomolar affinity and required the arginine at position 187 in a highly charged carboxy-terminal groove of the protein. In the absence of IFIT1, the growth and pathogenicity of viruses containing PPP-RNA was much greater. In contrast, IFIT proteins were dispensable for the clearance of pathogens that did not generate PPP-RNA. On the basis of this specificity and the great abundance of IFIT proteins after infection, we propose that the IFIT complex antagonizes viruses by sequestering specific viral nucleic acids.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue culture, and the human RSV leader region acts as a functional BRSV genome promoter.

            In order to generate recombinant bovine respiratory syncytial virus (BRSV), the genome of BRSV strain A51908, variant ATue51908, was cloned as cDNA. We provide here the sequence of the BRSV genome ends and of the entire L gene. This completes the sequence of the BRSV genome, which comprises a total of 15,140 nucleotides. To establish a vaccinia virus-free recovery system, a BHK-derived cell line stably expressing T7 RNA polymerase was generated (BSR T7/5). Recombinant BRSV was reproducibly recovered from cDNA constructs after T7 RNA polymerase-driven expression of antigenome sense RNA and of BRSV N, P, M2, and L proteins from transfected plasmids. Chimeric viruses in which the BRSV leader region was replaced by the human respiratory syncytial virus (HRSV) leader region replicated in cell culture as efficiently as their nonchimeric counterparts, demonstrating that all cis-acting sequences of the HRSV promoter are faithfully recognized by the BRSV polymerase complex. In addition, we report the successful recovery of a BRSV mutant lacking the complete NS2 gene, which encodes a nonstructural protein of unknown function. The NS2-deficient BRSV replicated autonomously and could be passaged, demonstrating that NS2 is not essential for virus replication in cell culture. However, growth of the mutant was considerably slower than and final infectious titers were reduced by a factor of at least 10 compared to wild-type BRSV, indicating that NS2 provides a supporting factor required for full replication capacity.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              5'-triphosphate RNA requires base-paired structures to activate antiviral signaling via RIG-I.

              The ATPase retinoid acid-inducible gene (RIG)-I senses viral RNA in the cytoplasm of infected cells and subsequently activates cellular antiviral defense mechanisms. RIG-I recognizes molecular structures that discriminate viral from host RNA. Here, we show that RIG-I ligands require base-paired structures in conjunction with a free 5'-triphosphate to trigger antiviral signaling. Hitherto unavailable chemically synthesized 5'-triphosphate RNA ligands do not trigger RIG-I-dependent IFN production in cells, and they are unable to trigger the ATPase activity of RIG-I without a base-paired stretch. Consistently, immunostimulatory RNA from cells infected with a virus recognized by RIG-I is sensitive to double-strand, but not single-strand, specific RNases. In vitro, base-paired stretches and the 5'-triphosphate bind to distinct sites of RIG-I and synergize to trigger the induction of signaling competent RIG-I multimers. Strengthening our model of a bipartite molecular pattern for RIG-I activation, we show that the activity of supposedly "single-stranded" 5'-triphosphate RNAs generated by in vitro transcription depends on extended and base-paired by-products inadvertently, but commonly, produced by this method. Together, our findings accurately define a minimal molecular pattern sufficient to activate RIG-I that can be found in viral genomes or transcripts.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                J Virol
                J. Virol
                jvi
                jvi
                JVI
                Journal of Virology
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                0022-538X
                1098-5514
                25 April 2018
                13 June 2018
                1 July 2018
                13 June 2018
                : 92
                : 13
                : e02140-17
                Affiliations
                [a ]MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
                [b ]Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, New York, USA
                [c ]Laboratory of Retrovirology, The Rockefeller University, New York, New York, USA
                [d ]Howard Hughes Medical Institute, The Rockefeller University, New York, New York, USA
                University of Southern California
                Author notes
                Address correspondence to Xiaohong Shi, Xiaohong.Shi@ 123456glasgow.ac.uk , or Sam J. Wilson, Sam.Wilson@ 123456glasgow.ac.uk .
                [*]

                Present address: Veronica V. Rezelj, Institut Pasteur, Viral Populations and Pathogenesis Unit, CNRS UMR 3569, Paris, France; Natasha L. Tilston-Lunel, Department of Microbiology/National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA; Gillian S. Slack, National Infection Service, Public Health England, Salisbury, United Kingdom.

                [†]

                Deceased.

                Citation Feng J, Wickenhagen A, Turnbull ML, Rezelj VV, Kreher F, Tilston-Lunel NL, Slack GS, Brennan B, Koudriakova E, Shaw AE, Rihn SJ, Rice CM, Bieniasz PD, Elliott RM, Shi X, Wilson SJ. 2018. Interferon-stimulated gene (ISG)-expression screening reveals the specific antibunyaviral activity of ISG20. J Virol 92:e02140-17. https://doi.org/10.1128/JVI.02140-17.

                Author information
                https://orcid.org/0000-0003-4241-5848
                https://orcid.org/0000-0003-4707-726X
                Article
                02140-17
                10.1128/JVI.02140-17
                6002717
                29695422
                49fb1d3c-6612-4de0-97a1-5627ddfa26d1
                Copyright © 2018 Feng et al.

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

                History
                : 20 December 2017
                : 14 April 2018
                Page count
                Figures: 6, Tables: 5, Equations: 0, References: 73, Pages: 20, Words: 12656
                Funding
                Funded by: Wellcome Trust;
                Award ID: 099220/Z/12/Z
                Award Recipient :
                Funded by: NIH NIAID;
                Award ID: R01-AI091707
                Award Recipient :
                Funded by: RCUK | Medical Research Council (MRC), https://doi.org/10.13039/501100000265;
                Award ID: MR/K024752/1
                Award ID: MC_UU_12014/10
                Award Recipient :
                Categories
                Virus-Cell Interactions
                Custom metadata
                July 2018

                Microbiology & Virology
                isg20,isgs,bunyavirus,innate immunity,interferons,restriction factor
                Microbiology & Virology
                isg20, isgs, bunyavirus, innate immunity, interferons, restriction factor

                Comments

                Comment on this article