16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Mutations in the Schmallenberg Virus Gc Glycoprotein Facilitate Cellular Protein Synthesis Shutoff and Restore Pathogenicity of NSs Deletion Mutants in Mice

      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

          Serial passage of viruses in cell culture has been traditionally used to attenuate virulence and identify determinants of viral pathogenesis. In a previous study, we found that a strain of Schmallenberg virus (SBV) serially passaged in tissue culture (termed SBVp32) unexpectedly displayed increased pathogenicity in suckling mice compared to wild-type SBV. In this study, we mapped the determinants of SBVp32 virulence to the viral genome M segment. SBVp32 virulence is associated with the capacity of this virus to reach high titers in the brains of experimentally infected suckling mice. We also found that the Gc glycoprotein, encoded by the M segment of SBVp32, facilitates host cell protein shutoff in vitro. Interestingly, while the M segment of SBVp32 is a virulence factor, we found that the S segment of the same virus confers by itself an attenuated phenotype to wild-type SBV, as it has lost the ability to block the innate immune system of the host. Single mutations present in the Gc glycoprotein of SBVp32 are sufficient to compensate for both the attenuated phenotype of the SBVp32 S segment and the attenuated phenotype of NSs deletion mutants. Our data also indicate that the SBVp32 M segment does not act as an interferon (IFN) antagonist. Therefore, SBV mutants can retain pathogenicity even when they are unable to fully control the production of IFN by infected cells. Overall, this study suggests that the viral glycoprotein of orthobunyaviruses can compensate, at least in part, for the function of NSs. In addition, we also provide evidence that the induction of total cellular protein shutoff by SBV is determined by multiple viral proteins, while the ability to control the production of IFN maps to the NSs protein.

          IMPORTANCE The identification of viral determinants of pathogenesis is key to the development of prophylactic and intervention measures. In this study, we found that the bunyavirus Gc glycoprotein is a virulence factor. Importantly, we show that mutations in the Gc glycoprotein can restore the pathogenicity of attenuated mutants resulting from deletions or mutations in the nonstructural protein NSs. Our findings highlight the fact that careful consideration should be taken when designing live attenuated vaccines based on deletions of nonstructural proteins since single mutations in the viral glycoproteins appear to revert attenuated mutants to virulent phenotypes.

          Related collections

          Most cited references26

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

          Novel Orthobunyavirus in Cattle, Europe, 2011

          In 2011, an unidentified disease in cattle was reported in Germany and the Netherlands. Clinical signs included fever, decreased milk production, and diarrhea. Metagenomic analysis identified a novel orthobunyavirus, which subsequently was isolated from blood of affected animals. Surveillance was initiated to test malformed newborn animals in the affected region.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Orthobunyaviruses: recent genetic and structural insights.

            Orthobunyaviruses, which have small, tripartite, negative-sense RNA genomes and structurally simple virions composed of just four proteins, can have devastating effects on human health and well-being, either by causing disease in humans or by causing disease in livestock and crops. In this Review, I describe the recent genetic and structural advances that have revealed important insights into the composition of orthobunyavirus virions, viral transcription and replication and viral interactions with the host innate immune response. Lastly, I highlight outstanding questions and areas of future research.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Culicoids as Vectors of Schmallenberg Virus

              To the Editor: In autumn 2011, an unidentified disease of livestock was reported on both sides of the Dutch–Germany border. By using metagenomics, the etiologic agent of this disease was identified as a novel orthobunyavirus and named Schmallenberg virus (SBV) ( 1 ). Other members of the genus Orthobunyavirus (e.g., Akabane virus) are widespread in Africa and Asia; biting midges (Culicoides spp.) and mosquitoes are responsible for transmitting these viruses. Hence, we reasonably assumed that European culicoids might be responsible for transmitting SBV within Europe. We present evidence that culicoids captured October 2011 in Denmark contained SBV RNA and most likely are vectors for this agent. In autumn 2011, culicoids were collected from several sites within Denmark. One site, a chicken farm in Hokkerup (Figure A1), was selected for study because of its location close (6 km) to the German border and proximity (<10 km) to an SBV-infected sheep farm in Germany, as reported on March 9, 2012, by the Friedrich Loeffler Institute surveillance website (www.fli.bund.de). The culicoids were collected during October 14–16 by using a Mosquito Magnet Independence trap (Mosquito Magnet, Lititz, PA, USA) baited with carbon dioxide and octenol. Midges were sorted manually into 91 specimens of the C. obsoletus group (comprising C. obsoletus, C. chiopterus, C. dewulfi, and C. scoticus) and 17 of the C. punctatus sensu stricto group, then stored at −20°C. Pools of culicoids were homogenized in water (100 µL) by using a 3-mm stainless steel bead (Dejay Distribution Ltd., Launceston, UK) in a TissueLyser II (QIAGEN, Hilden, Germany) for 1 min at 25 Hz ( 2 ). After homogenization, additional water (100 µL) was added to the samples, and then the mixture was centrifuged at 3,000 × g for 5 min. Nucleic acids were extracted from the supernatant (100 µL) by using a MagNA pure LC Total Nucleic Acid Isolation Kit on a MagNA pure LC (Roche Diagnostics, Basel, Switzerland) and eluted in water (50 µL). Two separate 1-step reverse transcription quantitative PCRs (RT-qPCRs), targeting the L segment and the S segment of SBV RNA, were performed according to protocols provided by the Friedrich Loeffler Institute in Germany ( 1 ) on the extracted nucleic acids by using a Mx3005p qPCR system (Agilent Technologies, Palo Alto, CA, USA). Another RT-qPCR targeting ruminant β-actin mRNA was performed as an internal endogenous control ( 3 ). Two of 22 pools tested strongly positive for the large (L) and small (S) segments of SBV RNA. Each positive sample was derived from 5 midges of the C. obsoletus group. One pool produced cycle threshold (Ct) values of 26.4 and 24.5 (in the L segment– and S segment–specific assays, respectively), whereas the second positive pool gave Ct values of 28.8 (L segment) and 27.6 (S segment). These pools were negative for the internal endogenous control that targeted the bovine/ovine β-actin mRNA. This result makes it unlikely that the detection of SBV RNA within the midges resulted from recent blood meals from infected animals remaining within the culicoids and suggests the virus has replicated within the midges. The PCR amplicons (145 bp; Figure) from the L segment–specific RT-qPCR were sequenced by using BigDye 1.1 chemistry on an ABI 3500 Genetic Analyzer (Applied Biosystems, Foster City, CA, USA). The sequences of 80 bp from the amplicons, excluding the primer sequences, had 100% identity with the expected region of the SBV segment L ( 1 ). Figure RNA extracted from pools of Culicoides obsoletus group midges was tested in 1-step reverse transcription quantitative PCRs (RT-qPCRs) for the Schmallenberg virus large segment, and the products were analyzed by agarose gel electrophoresis. Lanes 1–8, C. obsoletus group midge pools 1–8; lanes 9–10; negative and positive controls, respectively. Numbers below lanes are cycle threshold values from RT-qPCRs; –, no value. M, size marker. Amplicons (145 bp) from positive pools were extracted and sequenced. Reported Ct values generated by using the same assays from blood of naturally infected cattle were 24–35 ( 1 ). Usually, ≈100 µL of bovine/ovine blood is used for virus detection, whereas <1 µL of blood remains in a midge after a blood meal. This uptake of blood should therefore lead to a Ct value that is at least 6–7 units higher (≈100-fold lower level of RNA) when a single midge is tested by RT-qPCR ( 4 ). Thus, even if all 5 culicoids in a pool had recently taken a blood meal from a viremic animal, the Ct values observed here strongly suggest replication of SBV within the C. obsoletus group midges. However, in principle, other hosts of SBV could have a much higher level of viremia than cattle and could provide the levels of SBV RNA detected. C. punctatus s.s. midges cannot be ruled out as a possible vector of SBV because of the limited number of insects tested. Our study demonstrates the presence of SBV RNA in C. obsoletus group midges caught in Denmark during October 2011. The low Ct values (i.e., high SBV RNA levels) and the absence of ruminant β-actin mRNA in these samples strongly suggest that SBV replicates in these midges and hence that the C. obsoletus group midges are natural vectors for this virus.
                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
                16 March 2016
                12 May 2016
                1 June 2016
                12 May 2016
                : 90
                : 11
                : 5440-5450
                Affiliations
                [a ]MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
                [b ]Istituto Zooprofilattico Sperimentale dell'Abruzzo e Molise G. Caporale, Teramo, Italy
                [c ]Department of Pathology and Center of Systems Neuroscience, University of Veterinary Medicine, Hannover, Germany
                [d ]Virology Research Center, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirao Preto, Brazil
                Author notes
                Address correspondence to Mariana Varela, Mariana.varela@ 123456glasgow.ac.uk , or Massimo Palmarini, Massimo.palmarini@ 123456glasgow.ac.uk .
                [*]

                Present address: Rute Maria Pinto, The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom.

                Citation Varela M, Pinto RM, Caporale M, Piras IM, Taggart A, Seehusen F, Hahn K, Janowicz A, de Souza WM, Baumgärtner W, Shi X, Palmarini M. 2016. Mutations in the Schmallenberg virus Gc glycoprotein facilitate cellular protein synthesis shutoff and restore pathogenicity of NSs deletion mutants in mice. J Virol 90:5440–5450. doi: 10.1128/JVI.00424-16.

                Article
                00424-16
                10.1128/JVI.00424-16
                4934738
                26984728
                33908e7b-f3f0-4483-a05f-2061b6a4bca0
                Copyright © 2016 Varela et al.

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

                History
                : 2 March 2016
                : 15 March 2016
                Page count
                Figures: 9, Tables: 2, Equations: 0, References: 31, Pages: 11, Words: 8472
                Funding
                This work was funded by the Medical Research Council (MRC) and the Wellcome Trust. Funding was also provided by the Italian Ministry of Health (grant IZS A&M MSRCTE06.11), the Niedersachsen Research Network on Neuroinfectiology (N-RENNT) of the Ministry of Science and Culture of Lower Saxony, Germany, the Zoonotic Anticipation and Preparedness Initiative (Innovative Medicines Initiative grant 115760), and the Fundação de Amparo à Pesquisa do Estado de São Paulo, Brazil (scholarship number 15/05778-5).
                Categories
                Pathogenesis and Immunity

                Microbiology & Virology
                Microbiology & Virology

                Comments

                Comment on this article