1887

Abstract

Viperin has antiviral function against many viruses, including dengue virus (DENV), when studied in cells in culture. Here, the antiviral actions of viperin were defined both and in a mouse model of DENV infection. Murine embryonic fibroblasts (MEFs) derived from mice lacking viperin (vip) showed enhanced DENV infection, accompanied by increased IFN-β and induction of ISGs; IFIT1 and CXCL-10 but not IRF7, when compared to wild-type (WT) MEFs. In contrast, subcutaneous challenge of immunocompetent WT and vip mice with DENV did not result in enhanced infection. Intracranial infection with DENV resulted in body weight loss and neurological disease with a moderate increase in mortality in vip compared with WT mice, although this was not accompanied by altered brain morphology, immune cell infiltration or DENV RNA level in the brain. Similarly, DENV induction of IFN-β, IFIT1, CXCL-10, IRF7 and TNF-α was not significantly different in WT and vip mouse brain, although there was a modest but significant increase in DENV induction of IL-6 and IfI27la in the absence of viperin. NanoString nCounter analysis confirmed no significant difference in induction of a panel of inflammatory genes in WT compared to vip DENV-infected mouse brains. Further, polyI:C stimulation of bone marrow-derived macrophages (BMDMs) induced TNF-α, IFN-β, IL-6 and Nos-2, but responses were not different in BMDMs generated from WT or vip mice. Thus, while there is significant evidence of anti-DENV actions of viperin in some cell types , for DENV infection a lack of viperin does not affect systemic or brain susceptibility to DENV or induction of innate and inflammatory responses.

  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001669
2021-10-19
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/jgv/102/10/jgv001669.html?itemId=/content/journal/jgv/10.1099/jgv.0.001669&mimeType=html&fmt=ahah

References

  1. Ghosh S, Marsh ENG. Viperin: An ancient radical SAM enzyme finds its place in modern cellular metabolism and innate immunity. J Biol Chem 2020; 295:11513–11528 [View Article] [PubMed]
    [Google Scholar]
  2. Helbig KJ, Beard MR. The Role of Viperin in the Innate Antiviral Response. J Mol Biol 2014; 426:1210–1219 [View Article] [PubMed]
    [Google Scholar]
  3. Mattijssen S, Pruijn GJ. Viperin, a key player in the antiviral response. Microbes Infect 2012; 14:419–426 [View Article] [PubMed]
    [Google Scholar]
  4. Hinson ER, Cresswell P. The N-terminal amphipathic alpha-helix of viperin mediates localization to the cytosolic face of the endoplasmic reticulum and inhibits protein secretion. J Biol Chem 2009; 284:4705–4712 [View Article] [PubMed]
    [Google Scholar]
  5. Teng TS, Foo SS, Simamarta D, Lum FM, Teo TH et al. Viperin restricts chikungunya virus replication and pathology. J Clin Invest 2012; 122:4447–4460 [View Article] [PubMed]
    [Google Scholar]
  6. Wang X, Hinson ER, Cresswell P. The interferon-inducible protein viperin inhibits influenza virus release by perturbing lipid rafts. Cell Host Microbe 2007; 2:96–105 [View Article] [PubMed]
    [Google Scholar]
  7. Helbig KJ, Eyre NS, Yip E, Narayana S, Li K et al. The antiviral protein viperin inhibits hepatitis C virus replication via interaction with nonstructural protein 5A. Hepatology 2011; 54:1506–1517 [View Article] [PubMed]
    [Google Scholar]
  8. Van der Hoek KH, Eyre NS, Shue B, Khantisitthiporn O, Glab-Ampi K et al. Viperin is an important host restriction factor in control of Zika virus infection. Sci Rep 2017; 7:4475 [View Article] [PubMed]
    [Google Scholar]
  9. Upadhyay AS, Vonderstein K, Pichlmair A, Stehling O, Bennett KL et al. Viperin is an iron-sulfur protein that inhibits genome synthesis of tick-borne encephalitis virus via radical SAM domain activity. Cell Microbiol 2014; 16:834–848 [View Article] [PubMed]
    [Google Scholar]
  10. Jiang D, Weidner JM, Qing M, Pan XB, Guo H et al. Identification of five interferon-induced cellular proteins that inhibit west nile virus and dengue virus infections. J Virol 2010; 84:8332–8341 [View Article] [PubMed]
    [Google Scholar]
  11. Helbig KJ, Carr JM, Calvert JK, Wati S, Clarke JN et al. Viperin is induced following dengue virus type-2 (DENV-2) infection and has anti-viral actions requiring the C-terminal end of viperin. PLoS Negl Trop Dis 2013; 7:e2178 [View Article] [PubMed]
    [Google Scholar]
  12. Al-Shujairi WH, Clarke JN, Davies LT, Alsharifi M, Pitson SM et al. Intracranial Injection of Dengue Virus Induces Interferon Stimulated Genes and CD8+ T Cell Infiltration by Sphingosine Kinase 1 Independent Pathways. PLoS One 2017; 12:e0169814 [View Article] [PubMed]
    [Google Scholar]
  13. Norbury AJ, Calvert JK, Al-Shujairi WH, Cabezas-Falcon S, Tang V et al. Dengue virus infects the mouse eye following systemic or intracranial infection and induces inflammatory responses. J Gen Virol 2020; 101:79–85 [View Article] [PubMed]
    [Google Scholar]
  14. Szretter KJ, Brien JD, Thackray LB, Virgin HW, Cresswell P et al. The interferon-inducible gene viperin restricts West Nile virus pathogenesis. J Virol 2011; 85:11557–11566 [View Article] [PubMed]
    [Google Scholar]
  15. Tan KS, Olfat F, Phoon MC, Hsu JP, Howe JLC et al. In vivo and in vitro studies on the antiviral activities of viperin against influenza H1N1 virus infection. J Gen Virol 2012; 93:1269–1277 [View Article] [PubMed]
    [Google Scholar]
  16. Lindqvist R, Kurhade C, Gilthorpe JD, Överby AK. Cell-type- and region-specific restriction of neurotropic flavivirus infection by viperin. J Neuroinflammation 2018; 15:80 [View Article] [PubMed]
    [Google Scholar]
  17. Gualano RC, Pryor MJ, Cauchi MR, Wright PJ, Davidson AD. Identification of a major determinant of mouse neurovirulence of dengue virus type 2 using stably cloned genomic-length cDNA. J Gen Virol 1998; 79:437–446 [View Article] [PubMed]
    [Google Scholar]
  18. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008; 3:1101–1108 [View Article] [PubMed]
    [Google Scholar]
  19. Saitoh T, Satoh T, Yamamoto N, Uematsu S, Takeuchi O et al. Antiviral protein Viperin promotes Toll-like receptor 7- and Toll-like receptor 9-mediated type I interferon production in plasmacytoid dendritic cells. Immunity 2011; 34:352–363 [View Article] [PubMed]
    [Google Scholar]
  20. Johnson AJ, Roehrig JT. New mouse model for dengue virus vaccine testing. J Virol 1999; 73:783–786 [View Article] [PubMed]
    [Google Scholar]
  21. Orozco S, Schmid MA, Parameswaran P, Lachica R, Henn MR et al. Characterization of a model of lethal dengue virus 2 infection in C57BL/6 mice deficient in the alpha/beta interferon receptor. J Gen Virol 2012; 93:2152–2157 [View Article]
    [Google Scholar]
  22. Shresta S, Sharar KL, Prigozhin DM, Snider HM, Beatty PR et al. Critical roles for both STAT1-dependent and STAT1-independent pathways in the control of primary dengue virus infection in mice. J Immunol 2005; 175:3946–3954 [View Article] [PubMed]
    [Google Scholar]
  23. Tan GK, Ng JKW, Trasti SL, Schul W, Yip G et al. A non mouse-adapted dengue virus strain as a new model of severe dengue infection in AG129 mice. PLoS Negl Trop Dis 2010; 4:e672 [View Article] [PubMed]
    [Google Scholar]
  24. Cabezas-Falcon S, Norbury AJ, Hulme-Jones J, Klebe S, Adamson P et al. Changes in complement alternative pathway components, factor B and factor H during dengue virus infection in the AG129 mouse. J Gen Virol 2021; 102: [View Article]
    [Google Scholar]
  25. Al-Shujairi WH, Clarke JN, Davies LT, Pitman MR, Calvert JK et al. In vitro and in vivo roles of sphingosine kinase 2 during dengue virus infection. J Gen Virol 2019; 100:629–641 [View Article] [PubMed]
    [Google Scholar]
  26. Amaral DC, Rachid MA, Vilela MC, Campos RD, Ferreira GP et al. Intracerebral infection with dengue-3 virus induces meningoencephalitis and behavioral changes that precede lethality in mice. J Neuroinflammation 2011; 8:23 [View Article] [PubMed]
    [Google Scholar]
  27. de Miranda AS, Rodrigues DH, Amaral DC, de Lima Campos RD, Cisalpino D et al. Dengue-3 encephalitis promotes anxiety-like behavior in mice. Behav Brain Res 2012; 230:237–242 [View Article] [PubMed]
    [Google Scholar]
  28. Tsai TT, Chen CL, Lin YS, Chang CP, Tsai CC et al. Microglia retard dengue virus-induced acute viral encephalitis. Sci Rep 2016; 6:27670 [View Article] [PubMed]
    [Google Scholar]
  29. Lucas TM, Richner JM, Diamond MS. The interferon-stimulated gene Ifi27l2a restricts west nile virus infection and pathogenesis in a cell-type- and region-specific manner. J Virol 2015; 90:2600–2615 [View Article] [PubMed]
    [Google Scholar]
  30. Eom J, Yoo J, Kim JJ, Lee JB, Choi W et al. Viperin deficiency promotes polarization of macrophages and secretion of M1 and M2 cytokines. Immune Netw 2018; 18:e32 [View Article] [PubMed]
    [Google Scholar]
  31. Schneider WM, Chevillotte MD, Rice CM. Interferon-stimulated genes: a complex web of host defenses. Annu Rev Immunol 2014; 32:513–545 [View Article] [PubMed]
    [Google Scholar]
  32. Schoggins JW. Interferon-stimulated genes: roles in viral pathogenesis. Curr Opin Virol 2014; 6:40–46 [View Article] [PubMed]
    [Google Scholar]
  33. Schoggins JW, Rice CM. Interferon-stimulated genes and their antiviral effector functions. Curr Opin Virol 2011; 1:519–525 [View Article] [PubMed]
    [Google Scholar]
  34. Calvert JK, Helbig KJ, Dimasi D, Cockshell M, Beard MR et al. Dengue virus infection of primary endothelial cells induces innate immune responses, changes in endothelial cells function and is restricted by interferon-stimulated responses. J Interferon Cytokine Res 2015; 35:654–665 [View Article] [PubMed]
    [Google Scholar]
  35. Aloia AL, Calvert JK, Clarke JN, Davies LT, Helbig KJ et al. Investigation of sphingosine kinase 1 in interferon responses during dengue virus infection. Clin Transl Immunology 2017; 6:e151 [View Article] [PubMed]
    [Google Scholar]
  36. Nasr N, Maddocks S, Turville SG, Harman AN, Woolger N et al. HIV-1 infection of human macrophages directly induces viperin which inhibits viral production. Blood 2012; 120:778–788 [View Article] [PubMed]
    [Google Scholar]
  37. Tang YD, Na L, Zhu CH, Shen N, Yang F et al. Equine viperin restricts equine infectious anemia virus replication by inhibiting the production and/or release of viral Gag, Env, and receptor via distortion of the endoplasmic reticulum. J Virol 2014; 88:12296–12310 [View Article] [PubMed]
    [Google Scholar]
  38. Chan Y-L, Chang T-H, Liao C-L, Lin Y-L. The cellular antiviral protein viperin is attenuated by proteasome-mediated protein degradation in Japanese encephalitis virus-infected cells. J Virol 2008; 82:10455–10464 [View Article] [PubMed]
    [Google Scholar]
  39. Chan KW, Watanabe S, Kavishna R, Alonso S, Vasudevan SG. Animal models for studying dengue pathogenesis and therapy. Antiviral Res 2015; 123:5–14 [View Article] [PubMed]
    [Google Scholar]
  40. Zellweger RM, Shresta S. Mouse models to study dengue virus immunology and pathogenesis. Front Immunol 2014; 5:151 [View Article] [PubMed]
    [Google Scholar]
  41. Wei C, Zheng C, Sun J, Luo D, Tang Y et al. Viperin Inhibits Enterovirus A71 Replication by Interacting with Viral 2C Protein. Viruses 2018; 11:13 [View Article]
    [Google Scholar]
  42. Qiu LQ, Cresswell P, Chin KC. Viperin is required for optimal Th2 responses and T-cell receptor-mediated activation of NF-kappaB and AP-1. Blood 2009; 113:3520–3529 [View Article] [PubMed]
    [Google Scholar]
  43. Cho H, Proll SC, Szretter KJ, Katze MG, Gale M et al. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat Med 2013; 19:458–464 [View Article] [PubMed]
    [Google Scholar]
  44. Ebrahimi KH, Howie D, Rowbotham JS, McCullagh J, Armstrong FA et al. Viperin, through its radical-SAM activity, depletes cellular nucleotide pools and interferes with mitochondrial metabolism to inhibit viral replication. FEBS Lett 2020; 594:1624–1630 [View Article] [PubMed]
    [Google Scholar]
  45. Clarke P, Leser JS, Bowen RA, Tyler KL. Virus-induced transcriptional changes in the brain include the differential expression of genes associated with interferon, apoptosis, interleukin 17 receptor A, and glutamate signaling as well as flavivirus-specific upregulation of tRNA synthetases. mBio 2014; 5:e00902–14 [View Article] [PubMed]
    [Google Scholar]
  46. Wu J, Chen ZJ. Innate immune sensing and signaling of cytosolic nucleic acids. Annu Rev Immunol 2014; 32:461–488 [View Article] [PubMed]
    [Google Scholar]
  47. Eom J, Kim JJ, Yoon SG, Jeong H, Son S et al. Intrinsic expression of viperin regulates thermogenesis in adipose tissues. Proc Natl Acad Sci U S A 2019; 116:17419–17428 [View Article] [PubMed]
    [Google Scholar]
  48. Steinbusch MMF, Caron MMJ, Surtel DAM, van den Akker GGH, van Dijk PJ et al. The antiviral protein viperin regulates chondrogenic differentiation via CXCL10 protein secretion. J Biol Chem 2019; 294:5121–5136 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001669
Loading
/content/journal/jgv/10.1099/jgv.0.001669
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Supplementary material 2

EXCEL
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error