Message in a bottle: lessons learned from antagonism of STING signalling during RNA virus infection

Kevin Maringer, Ana Fernandez-Sesma, Kevin Maringer, Ana Fernandez-Sesma

Abstract

STING has emerged in recent years as an important signalling adaptor in the activation of type I interferon responses during infection with DNA viruses and bacteria. An increasing body of evidence suggests that STING also modulates responses to RNA viruses, though the mechanisms remain less clear. In this review, we give a brief overview of the ways in which STING facilitates sensing of RNA viruses. These include modulation of RIG-I-dependent responses through STING's interaction with MAVS, and more speculative mechanisms involving the DNA sensor cGAS and sensing of membrane remodelling events. We then provide an in-depth literature review to summarise the known mechanisms by which RNA viruses of the families Flaviviridae and Coronaviridae evade sensing through STING. Our own work has shown that the NS2B/3 protease complex of the flavivirus dengue virus binds and cleaves STING, and that an inability to degrade murine STING may contribute to host restriction in this virus. We contrast this to the mechanism employed by the distantly related hepacivirus hepatitis C virus, in which STING is bound and inactivated by the NS4B protein. Finally, we discuss STING antagonism in the coronaviruses SARS coronavirus and human coronavirus NL63, which disrupt K63-linked polyubiquitination and dimerisation of STING (both of which are required for STING-mediated activation of IRF-3) via their papain-like proteases. We draw parallels with less-well characterised mechanisms of STING antagonism in related viruses, and place our current knowledge in the context of species tropism restrictions that potentially affect the emergence of new human pathogens.

Keywords: Dengue; Hepatitis C virus; Immune evasion; SARS coronavirus; STING.

Copyright © 2014 The Authors. Published by Elsevier Ltd.. All rights reserved.

Figures

Fig. 1
Fig. 1
STING signalling during RNA virus infection. Inactive STING resides in membranes of the ER, MAM and mitochondria (M*). Following its activation, STING dimerises and relocalises to perinuclear punctate domains, where it interacts with TBK1 to phosphorylate IRF-3. Activated STING is also modified by phosphorylation and K63-linked polyubiquitination. Following its activation, IRF-3 dimerises and translocates to the nucleus, where it induces the transcription of the type I interferons IFN-α and IFN-β. RNA viruses have been proposed to be sensed by three distinct mechanisms in relation to STING activation. (1) Viral dsRNA replication intermediates and stem-loop structures, and the 5′-triphosphates of uncapped viral RNAs are recognised by RIG-I, which associates with MAVS and STING at MAMs to activate STING. (2) The DNA sensor cGAS has been linked to the sensing of positive-sense RNA viruses by an as-yet undefined mechanism. In this scenario, cGAS produces the messenger molecule cGAMP after detecting RNA virus infection, and cGAMP binds and activates STING. (3) Viral fusion events at the plasma membrane have been shown to activate STING by a poorly-defined mechanism involving PI(3)K and phospholipase C-γ (PLC-γ). For simplicity, only signalling molecules referred to in the main text are shown.
Fig. 2
Fig. 2
Flaviviridae phylogenetic tree and genome organisation. (A) Flaviviridae family tree. Viruses of relevance to this review, and other representative viruses, shown. (B) Mosquito-borne flavivirus (∼11 kb) and (C) hepacivirus (∼9.6 kb) genome organisation (not to scale). Structural proteins are in black. STING antagonists are highlighted in grey. A STING antagonist function has been described for the YFV NS4B protein, but not for the DENV NS4B. Additional known functions for proteins discussed in this review are also given. Triangles indicate cleavage sites for the flavivirus NS2B/3 (B) or hepacivirus NS3/4A (C) proteases. The HCV polyuridine tract ((U/UC)n) is a well-known PAMP that induces type I IFN production. Note that the hepacivirus genome is uncapped. Members of the Flaviviridae lack a poly(A) tail. Viruses not defined elsewhere; CSFV, classical swine fever virus; BDV, bovine diarrhoea virus; GBV, GV virus.
Fig. 3
Fig. 3
Coronavirinae phylogenetic tree, genome organisation, and nsp3 functional domains. (A) Coronavirinae family tree. Viruses of relevance to this review, and other representative viruses, shown. (B) SARS-CoV genome organisation (not to scale). Structural proteins are in black. The two polyproteins (1a and 1ab) translated by ribosomal frameshifting are shown. Triangles indicate PLpro cleavage sites; the remaining polyprotein cleavage sites are processed by the main 3C-like protease. The 3′ structural and additional accessory proteins differ among coronaviruses. Functional domains encoded by the SARS-CoV (C) and the HCoV-NL63 (D) nsp3 are also shown (not to scale). Domains linked to STING antagonism are highlighted in grey; the precise function of the transmembrane (TM) domain remains unknown. Terms not defined elsewhere; TCoV, turkey coronavirus; IBV, infectious bronchitis virus; Ubl, ubiquitin-like domain; Ac, acidic domain; ADRP, poly(ADP-ribose)-binding/ADP-ribose-1′-phosphatase macrodomain; NAB, nucleic acid-binding domain; G2M, betacoronavirus marker; Y domain, highly conserved coronavirus domain.

References

    1. Yoneyama M., Fujita T. RIG-I family RNA helicases: cytoplasmic sensor for antiviral innate immunity. Cytokine Growth Factor Rev. 2007;18:545–551.
    1. Garcia-Sastre A., Biron C.A. Type 1 interferons and the virus–host relationship: a lesson in detente. Sci Rep. 2006;312:879–882.
    1. Ishikawa H., Barber G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674–678.
    1. Zhong B., Yang Y., Li S., Wang Y.-Y., Li Y., Diao F. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity. 2008;29:538–550.
    1. Sun W., Li Y., Chen L., Chen H., You F., Zhou X. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proc Natl Acad Sci USA. 2009;106:8653–8658.
    1. Unterholzner L. The interferon response to intracellular DNA: why so many receptors? Immunobiology. 2013;218:1312–1321.
    1. Ran Y., Shu H.-B., Wang Y.-Y. MITA/STING: a central and multifaceted mediator in innate immune response. Cytokine Growth Factor Rev. 2014:1–9.
    1. Ishikawa H., Ma Z., Barber G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature. 2009;461:788–792.
    1. Aguirre S., Maestre A.M., Pagni S., Patel J.R., Savage T., Gutman D. DENV inhibits type I IFN production in infected cells by cleaving human STING. PLoS Pathog. 2012;8:e1002934.
    1. Nitta S., Sakamoto N., Nakagawa M., Kakinuma S., Mishima K., Kusano-Kitazume A. Hepatitis C virus NS4B protein targets STING and abrogates RIG-I-mediated type I interferon-dependent innate immunity. Hepatology. 2013;57:46–58.
    1. Sun L., Xing Y., Chen X., Zheng Y., Yang Y., Nichols D.B. Coronavirus papain-like proteases negatively regulate antiviral innate immune response through disruption of STING-mediated signaling. PLoS ONE. 2012;7:e30802.
    1. Yu C.-Y., Chang T.-H., Liang J.-J., Chiang R.-L., Lee Y.-L., Liao C.-L. Dengue virus targets the adaptor protein MITA to subvert host innate immunity. PLoS Pathog. 2012;8:e1002780.
    1. Ding Q., Cao X., Lu J., Huang B., Liu Y.-J., Kato N. Hepatitis C virus NS4B blocks the interaction of STING and TBK1 to evade host innate immunity. J Hepatol. 2013;59:52–58.
    1. Chen X., Yang X., Zheng Y., Yang Y., Xing Y., Chen Z. SARS coronavirus papain-like protease inhibits the type I interferon signaling pathway through interaction with the STING-TRAF3-TBK1 complex. Protein Cell. 2014;5:369–381.
    1. Xing Y., Chen J., Tu J., Zhang B., Chen X., Shi H. The papain-like protease of porcine epidemic diarrhea virus negatively regulates type I interferon pathway by acting as a viral deubiquitinase. J Gen Virol. 2013;94:1554–1567.
    1. Ouyang S., Song X., Wang Y., Ru H., Shaw N., Jiang Y. Structural analysis of the STING adaptor protein reveals a hydrophobic dimer interface and mode of cyclic di-GMP binding. Immunity. 2012;36:1073–1086.
    1. Tsuchida T., Zou J., Saitoh T., Kumar H., Abe T., Matsuura Y. The ubiquitin ligase TRIM56 regulates innate immune responses to intracellular double-stranded DNA. Immunity. 2010;33:765–776.
    1. Zhang J., Hu M.-M., Wang Y.-Y., Shu H.-B. TRIM32 protein modulates type I interferon induction and cellular antiviral response by targeting MITA/STING protein for K63-linked ubiquitination. J Biol Chem. 2012;287:28646–28655.
    1. Abe T., Harashima A., Xia T., Konno H., Konno K., Morales A. STING recognition of cytoplasmic DNA instigates cellular defense. Mol Cell. 2013;50:5–15.
    1. Bowzard J.B., Ranjan P., Sambhara S., Fujita T. Antiviral defense: RIG-Ing the immune system to STING. Cytokine Growth Factor Rev. 2009;20:1–5.
    1. Yoneyama M., Kikuchi M., Natsukawa T., Shinobu N., Imaizumi T., Miyagishi M. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:730–737.
    1. Schoggins J.W., MacDuff D.A., Imanaka N., Gainey M.D., Shrestha B., Eitson J.L. Pan-viral specificity of IFN-induced genes reveals new roles for cGAS in innate immunity. Nature. 2014;505:691–695.
    1. Schoggins J.W., Wilson S.J., Panis M., Murphy M.Y., Jones C.T., Bieniasz P. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485.
    1. Holm C.K., Jensen S.B., Jakobsen M.R., Cheshenko N., Horan K.A., Moeller H.B. Virus-cell fusion as a trigger of innate immunity dependent on the adaptor STING. Nat Immunol. 2012;13:737–743.
    1. Sun L., Wu J., Du F., Chen X., Chen Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Sci Rep. 2013;339:786–791.
    1. Civril F., Deimling T., de Oliveira Mann C.C., Ablasser A., Moldt M., Witte G. Structural mechanism of cytosolic DNA sensing by cGAS. Nature. 2013;498:332–337.
    1. Paul D., Bartenschlager R. Architecture and biogenesis of plus-strand RNA virus replication factories. World J Virol. 2013;2:32–48.
    1. Lindenbach B.D., Thiel H.-J., Rice C.M. Flaviviridae: the viruses and their replication. In: Knipe D.M., Howley P.M., editors. 5th ed. vol. I. Lippincott Williams and Wilkins; Philadelphia, PA USA: 2007. pp. 1102–1152. (Fields virology).
    1. Clyde K., Kyle J.L., Harris E. Recent advances in deciphering viral and host determinants of dengue virus replication and pathogenesis. J Virol. 2006;80:11418–11431.
    1. Mackenzie J.S., Gubler D.J., Petersen L.R. Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat Med. 2004;10:S98–S109.
    1. Heinz F.X., Stiasny K. Flaviviruses and flavivirus vaccines. Vaccine. 2012;30:4301–4306.
    1. Bhatt S., Gething P.W., Brady O.J., Messina J.P., Farlow A.W., Moyes C.L. The global distribution and burden of dengue. Nature. 2013;496:504–507.
    1. Weaver S.C., Barrett A.D.T. Transmission cycles, host range, evolution and emergence of arboviral disease. Nat Rev Microbiol. 2004;2:789–801.
    1. Gubler D.J. Dengue and dengue hemorrhagic fever. Clin Microbiol Rev. 1998;11:480–496.
    1. Halstead S.B. Dengue vaccine development: a 75% solution? Lancet. 2012;380:1535–1536.
    1. Thisyakorn U., Thisyakorn C. Latest developments and future directions in dengue vaccines. Ther Adv Vaccines. 2014;2:3–9.
    1. McBride W.J.H., Bielefeldt-Ohmann H. Dengue viral infections pathogenesis and epidemiology. Microbes Infect. 2000;2:1041–1050.
    1. Rodriguez-Madoz J.R., Bernal-Rubio D., Kaminski D., Boyd K., Fernandez-Sesma A. Dengue virus inhibits the production of type I interferon in primary human dendritic cells. J Virol. 2010;84:4845–4850.
    1. Rodriguez-Madoz J.R., Belicha-Villanueva A., Bernal-Rubio D., Ashour J., Ayllon J., Fernandez-Sesma A. Inhibition of the type I interferon response in human dendritic cells by dengue virus infection requires a catalytically active NS2B3 complex. J Virol. 2010;84:9760–9774.
    1. Jin L., Xu L.-G., Yang I.V., Davidson E.J., Schwartz D.A., Wurfel M.M. Identification and characterization of a loss-of-function human MPYS variant. Genes Immun. 2011;12:263–269.
    1. Meiklejohn G., England B., Lennette E. Propagation of dengue virus strains in unweaned mice. Am J Trop Med Hyg. 1952;1:51–58.
    1. Zompi S., Harris E. Animal models of dengue virus infection. Viruses. 2012;4:62–82.
    1. Ashour J., Morrison J., Laurent-Rolle M., Belicha-Villanueva A., Plumlee C.R., Bernal-Rubio D. Mouse STAT2 restricts early dengue virus replication. Cell Host Microbe. 2010;8:410–421.
    1. Shresta S., Sharar K.L., Prigozhin D.M., Beatty P.R., Harris E. Murine model for dengue virus-induced lethal disease with increased vascular permeability. J Virol. 2006;80:10208–10217.
    1. Chambers T.J., Hahn C.S., Galler R., Rice C.M. Flavivirus genome organization, expression, and replication. Annu Rev Microbiol. 1990;44:649–688.
    1. Shiryaev S.A., Kozlov I.A., Ratnikov B.I., Smith J.W., Lebl M., Strongin A.Y. Cleavage preference distinguishes the two-component NS2B–NS3 serine proteinases of Dengue and West Nile viruses. Biochem J. 2007;401:743.
    1. Nazmi A., Mukhopadhyay R., Dutta K., Basu A. STING mediates neuronal innate immune response following Japanese encephalitis virus infection. Sci Rep. 2012;2:1–10.
    1. Feng X., Yang C., Zhang Y., Peng L., Chen X., Rao Y. Identification, characterization and immunological response analysis of stimulator of interferon gene (STING) from grass carp Ctenopharyngodon idella. Dev Comp Immunol. 2014;45:163–176.
    1. Xie L., Liu M., Fang L., Su X., Cai K., Wang D. Molecular cloning and functional characterization of porcine stimulator of interferon genes (STING) Dev Comp Immunol. 2010;34:847–854.
    1. Kemp C., Imler J.-L. Antiviral immunity in drosophila. Curr Opin Immunol. 2009;21:3–9.
    1. Lavanchy D. The global burden of hepatitis C. Liver Int. 2009;29:74–81.
    1. Horner S.M. Activation and evasion of antiviral innate immunity by hepatitis C virus. J Mol Biol. 2014;426:1198–1209.
    1. Lemon S.M., Walker C., Alter M.J., Yi M. Hepatitis C virus. In: Knipe D.M., Howley P.M., editors. 5th ed. vol. I. Lippincott Williams and Wilkins; Philadelphia, PA USA: 2007. pp. 1254–1305. (Fields virology).
    1. Park S.-H., Rehermann B. Immune responses to HCV and other hepatitis viruses. Immunity. 2014;40:13–24.
    1. Liang T.J. Current progress in development of hepatitis C virus vaccines. Nat Med. 2013;19:869–878.
    1. Cheng G., Zhong J., Chisari F.V. Inhibition of dsRNA-induced signaling in hepatitis C virus-infected cells by NS3 protease-dependent and -independent mechanisms. Proc Natl Acad Sci USA. 2006;103:8499–8504.
    1. Moriyama M., Kato N., Otsuka M., Shao R.-X., Taniguchi H., Kawabe T. Interferon-beta is activated by hepatitis C virus NS5B and inhibited by NS4A, NS4B, and NS5A. Hepatol Int. 2007;1:302–310.
    1. Tasaka M., Sakamoto N., Itakura Y., Nakagawa M., Itsui Y., Sekine-Osajima Y. Hepatitis C virus non-structural proteins responsible for suppression of the RIG-I/Cardif-induced interferon response. J Gen Virol. 2007;88:3323–3333.
    1. Meylan E., Curran J., Hofmann K., Moradpour D., Binder M., Bartenschlager R. Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature. 2005;437:1167–1172.
    1. Baril M., Racine M.-E., Penin F., Lamarre D. MAVS dimer is a crucial signaling component of innate immunity and the target of hepatitis C virus NS3/4A protease. J Virol. 2009;83:1299–1311.
    1. Li X.-D., Sun L., Seth R.B., Pineda G., Chen Z.J. Hepatitis C virus protease NS3/4A cleaves mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc Natl Acad Sci USA. 2005;102:17717–17722.
    1. Saito T., Owen D.M., Jiang F., Marcotrigiano J., Gale M. Innate immunity induced by composition-dependent RIG-I recognition of hepatitis C virus RNA. Nature. 2008;454:523–527.
    1. Rai R., Deval J. New opportunities in anti-hepatitis C virus drug discovery: targeting NS4B. Antivir Res. 2011;90:93–101.
    1. Masters P.S., Perlman S. Coronaviridae. In: Knipe D.M., Howley P.M., editors. 6th ed. vol. I. Lippincott Williams and Wilkins; Philadelphia, PA USA: 2013. pp. 826–859. (Fields virology).
    1. Coleman C.M., Frieman M.B. Coronaviruses: important emerging human pathogens. J Virol. 2014;88:5209–5212.
    1. Davidson A.D., Siddell S. Potential for antiviral treatment of severe acute respiratory syndrome. Curr Opin Infect Dis. 2003;16:565–571.
    1. Kindler E., Thiel V. To sense or not to sense viral RNA – essentials of coronavirus innate immune evasion. Curr Opin Microbiol. 2014;20:69–75.
    1. Perlman S., Netland J. Coronaviruses post-SARS: update on replication and pathogenesis. Nat Rev Microbiol. 2009;7:439–450.
    1. Zhao J., Zhao J., Perlman S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J Virol. 2010;84:9318–9325.
    1. Devaraj S.G., Wang N., Chen Z., Chen Z., Tseng M., Barretto N. Regulation of IRF-3-dependent innate immunity by the papain-like protease domain of the severe acute respiratory syndrome coronavirus. J Biol Chem. 2007;282:32208–32221.
    1. Clementz M.A., Chen Z., Banach B.S., Wang Y., Sun L., Ratia K. Deubiquitinating and interferon antagonism activities of coronavirus papain-like proteases. J Virol. 2010;84:4619–4629.
    1. Ratia K., Saikatendu K.S., Santarsiero B.D., Barretto N., Baker S.C., Stevens R.C. Severe acute respiratory syndrome coronavirus papain-like protease: structure of a viral deubiquitinating enzyme. Proc Natl Acad Sci USA. 2006;103:5717–5722.
    1. Lindner H.A., Fotouhi-Ardakani N., Lytvyn V., Lachance P., Sulea T., Ménard R. The papain-like protease from the severe acute respiratory syndrome coronavirus is a deubiquitinating enzyme. J Virol. 2005;79:15199–15208.
    1. Lindner H.A., Lytvyn V., Qi H., Lachance P., Ziomek E., Ménard R. Selectivity in ISG15 and ubiquitin recognition by the SARS coronavirus papain-like protease. Arch Biochem Biophys. 2007;466:8–14.
    1. Barretto N., Jukneliene D., Ratia K., Chen Z., Mesecar A.D., Baker S.C. The papain-like protease of severe acute respiratory syndrome coronavirus has deubiquitinating activity. J Virol. 2005;79:15189–15198.
    1. Yang X., Chen X., Bian G., Tu J., Xing Y., Wang Y. Proteolytic processing, deubiquitinase and interferon antagonist activities of Middle East respiratory syndrome coronavirus papain-like protease. J Gen Virol. 2014;95:614–626.
    1. Zheng D., Chen G., Guo B., Cheng G., Tang H. PLP2, a potent deubiquitinase from murine hepatitis virus, strongly inhibits cellular type I interferon production. Cell Res. 2008;18:1105–1113.
    1. Wang G., Chen G., Zheng D., Cheng G., Tang H. PLP2 of mouse hepatitis virus A59 (MHV-A59) targets TBK1 to negatively regulate cellular type I interferon signaling pathway. PLoS ONE. 2011;6:e17192.

Source: PubMed

Подписаться