1887

Abstract

Interferon (IFN) regulatory factors (IRFs) are important determinants of the innate response to infection. We evaluated the role(s) of combined and individual IRF deficiencies in the outcome of infection of C57BL/6 mice with Sindbis virus, an alphavirus that infects neurons and causes encephalomyelitis. The brain and spinal cord levels of Irf7, but not Irf3 mRNAs, were increased after infection. IRF3/5/7−/− and IRF3/7−/− mice died within 3–4 days with uncontrolled virus replication, similar to IFNα receptor-deficient mice, while all wild-type (WT) mice recovered. IRF3−/− and IRF7−/− mice had brain levels of IFNα that were lower, but brain and spinal cord levels of IFNβ and IFN-stimulated gene mRNAs that were similar to or higher than WT mice without detectable serum IFN or increases in Ifna or Ifnb mRNAs in the lymph nodes, indicating that the differences in outcome were not due to deficiencies in the central nervous system (CNS) type I IFN response. IRF3−/− mice developed persistent neurological deficits and had more spinal cord inflammation and higher CNS levels of Il1b and Ifnγ mRNAs than WT mice, but all mice survived. IRF7−/− mice died 5–8 days after infection with rapidly progressive paralysis and differed from both WT and IRF3−/− mice in the induction of higher CNS levels of IFNβ, tumour necrosis factor (TNF) α and Cxcl13 mRNA, delayed virus clearance and more extensive cell death. Therefore, fatal disease in IRF7−/− mice is likely due to immune-mediated neurotoxicity associated with failure to regulate the production of inflammatory cytokines such as TNFα in the CNS.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001174
2018-11-19
2024-03-29
Loading full text...

Full text loading...

/deliver/fulltext/jgv/100/1/46.html?itemId=/content/journal/jgv/10.1099/jgv.0.001174&mimeType=html&fmt=ahah

References

  1. Gubler DJ. The global emergence/resurgence of arboviral diseases as public health problems. Arch Med Res 2002; 33:330–342 [View Article][PubMed]
    [Google Scholar]
  2. Griffin DE. Emergence and re-emergence of viral diseases of the central nervous system. Prog Neurobiol 2010; 91:95–101 [View Article][PubMed]
    [Google Scholar]
  3. Griffin DE. Role of the immune response in age-dependent resistance of mice to encephalitis due to Sindbis virus. J Infect Dis 1976; 133:456–464 [View Article][PubMed]
    [Google Scholar]
  4. Jackson AC, Moench TR, Trapp BD, Griffin DE. Basis of neurovirulence in Sindbis virus encephalomyelitis of mice. Lab Invest 1988; 58:503–509[PubMed]
    [Google Scholar]
  5. Schilte C, Couderc T, Chretien F, Sourisseau M, Gangneux N et al. Type I IFN controls chikungunya virus via its action on nonhematopoietic cells. J Exp Med 2010; 207:429–442 [View Article][PubMed]
    [Google Scholar]
  6. Ryman KD, Meier KC, Gardner CL, Adegboyega PA, Klimstra WB. Non-pathogenic Sindbis virus causes hemorrhagic fever in the absence of alpha/beta and gamma interferons. Virology 2007; 368:273–285 [View Article][PubMed]
    [Google Scholar]
  7. Ryman KD, Klimstra WB, Nguyen KB, Biron CA, Johnston RE. Alpha/beta interferon protects adult mice from fatal Sindbis virus infection and is an important determinant of cell and tissue tropism. J Virol 2000; 74:3366–3378 [View Article][PubMed]
    [Google Scholar]
  8. Fragkoudis R, Breakwell L, Mckimmie C, Boyd A, Barry G et al. The type I interferon system protects mice from Semliki Forest virus by preventing widespread virus dissemination in extraneural tissues, but does not mediate the restricted replication of avirulent virus in central nervous system neurons. J Gen Virol 2007; 88:3373–3384 [View Article][PubMed]
    [Google Scholar]
  9. Byrnes AP, Durbin JE, Griffin DE. Control of Sindbis virus infection by antibody in interferon-deficient mice. J Virol 2000; 74:3905–3908 [View Article][PubMed]
    [Google Scholar]
  10. Ikushima H, Negishi H, Taniguchi T. The IRF family transcription factors at the interface of innate and adaptive immune responses. Cold Spring Harb Symp Quant Biol 2013; 78:105–116 [View Article][PubMed]
    [Google Scholar]
  11. Tarassishin L, Bauman A, Suh HS, Lee SC. Anti-viral and anti-inflammatory mechanisms of the innate immune transcription factor interferon regulatory factor 3: relevance to human CNS diseases. J Neuroimmune Pharmacol 2013; 8:132–144 [View Article][PubMed]
    [Google Scholar]
  12. Yoneyama M, Suhara W, Fukuhara Y, Fukuda M, Nishida E et al. Direct triggering of the type I interferon system by virus infection: activation of a transcription factor complex containing IRF-3 and CBP/p300. EMBO J 1998; 17:1087–1095 [View Article][PubMed]
    [Google Scholar]
  13. Andersen J, Vanscoy S, Cheng TF, Gomez D, Reich NC. IRF-3-dependent and augmented target genes during viral infection. Genes Immun 2008; 9:168–175 [View Article][PubMed]
    [Google Scholar]
  14. Hiscott J. Triggering the innate antiviral response through IRF-3 activation. J Biol Chem 2007; 282:15325–15329 [View Article][PubMed]
    [Google Scholar]
  15. Marié I, Durbin JE, Levy DE. Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon regulatory factor-7. EMBO J 1998; 17:6660–6669 [View Article][PubMed]
    [Google Scholar]
  16. Sato M, Suemori H, Hata N, Asagiri M, Ogasawara K et al. Distinct and essential roles of transcription factors IRF-3 and IRF-7 in response to viruses for IFN-α/β gene induction. Immunity 2000; 13:539–548 [View Article][PubMed]
    [Google Scholar]
  17. Osterlund PI, Pietilä TE, Veckman V, Kotenko SV, Julkunen I. IFN regulatory factor family members differentially regulate the expression of type III IFN (IFN-λ) genes. J Immunol 2007; 179:3434–3442 [View Article][PubMed]
    [Google Scholar]
  18. Prakash A, Smith E, Lee CK, Levy DE. Tissue-specific positive feedback requirements for production of type I interferon following virus infection. J Biol Chem 2005; 280:18651–18657 [View Article][PubMed]
    [Google Scholar]
  19. Kawai T, Sato S, Ishii KJ, Coban C, Hemmi H et al. Interferon-α induction through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6. Nat Immunol 2004; 5:1061–1068 [View Article][PubMed]
    [Google Scholar]
  20. Ning S, Huye LE, Pagano JS. Regulation of the transcriptional activity of the IRF7 promoter by a pathway independent of interferon signaling. J Biol Chem 2005; 280:12262–12270 [View Article][PubMed]
    [Google Scholar]
  21. Marié I, Smith E, Prakash A, Levy DE. Phosphorylation-induced dimerization of interferon regulatory factor 7 unmasks DNA binding and a bipartite transactivation domain. Mol Cell Biol 2000; 20:8803–8814 [View Article][PubMed]
    [Google Scholar]
  22. Au WC, Moore PA, Lafleur DW, Tombal B, Pitha PM. Characterization of the interferon regulatory factor-7 and its potential role in the transcription activation of interferon A genes. J Biol Chem 1998; 273:29210–29217[PubMed]
    [Google Scholar]
  23. Zhang L, Pagano JS. IRF-7, a new interferon regulatory factor associated with Epstein-Barr virus latency. Mol Cell Biol 1997; 17:5748–5757 [View Article][PubMed]
    [Google Scholar]
  24. Lazear HM, Lancaster A, Wilkins C, Suthar MS, Huang A et al. IRF-3, IRF-5, and IRF-7 coordinately regulate the type I IFN response in myeloid dendritic cells downstream of MAVS signaling. PLoS Pathog 2013; 9:e1003118 [View Article][PubMed]
    [Google Scholar]
  25. Proenca-Modena JL, Hyde JL, Sesti-Costa R, Lucas T, Pinto AK et al. Interferon-regulatory factor 5-dependent signaling restricts orthobunyavirus dissemination to the central nervous system. J Virol 2015; 90:189–205 [View Article][PubMed]
    [Google Scholar]
  26. Gardner J, Rudd PA, Prow NA, Belarbi E, Roques P et al. Infectious Chikungunya virus in the saliva of mice, monkeys and humans. PLoS One 2015; 10:e0139481 [View Article][PubMed]
    [Google Scholar]
  27. Rudd PA, Wilson J, Gardner J, Larcher T, Babarit C et al. Interferon response factors 3 and 7 protect against Chikungunya virus hemorrhagic fever and shock. J Virol 2012; 86:9888–9898 [View Article][PubMed]
    [Google Scholar]
  28. Daffis S, Suthar MS, Szretter KJ, Gale M, Diamond MS. Induction of IFN-β and the innate antiviral response in myeloid cells occurs through an IPS-1-dependent signal that does not require IRF-3 and IRF-7. PLoS Pathog 2009; 5:e1000607 [View Article][PubMed]
    [Google Scholar]
  29. Honda K, Yanai H, Negishi H, Asagiri M, Sato M et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 2005; 434:772–777 [View Article][PubMed]
    [Google Scholar]
  30. Menachery VD, Pasieka TJ, Leib DA. Interferon regulatory factor 3-dependent pathways are critical for control of herpes simplex virus type 1 central nervous system infection. J Virol 2010; 84:9685–9694 [View Article][PubMed]
    [Google Scholar]
  31. Daffis S, Samuel MA, Keller BC, Gale M, Diamond MS. Cell-specific IRF-3 responses protect against West Nile virus infection by interferon-dependent and -independent mechanisms. PLoS Pathog 2007; 3:e106 [View Article][PubMed]
    [Google Scholar]
  32. Daffis S, Samuel MA, Suthar MS, Keller BC, Gale M et al. Interferon regulatory factor IRF-7 induces the antiviral alpha interferon response and protects against lethal West Nile virus infection. J Virol 2008; 82:8465–8475 [View Article][PubMed]
    [Google Scholar]
  33. Nilaratanakul V, Chen J, Tran O, Baxter VK, Troisi EM et al. Germ line IgM is sufficient, but not required, for antibody-mediated alphavirus clearance from the central nervous system. J Virol 2018; 92:e02081-17 [View Article][PubMed]
    [Google Scholar]
  34. Baxter VK, Troisi EM, Pate NM, Zhao JN, Griffin DE. Death and gastrointestinal bleeding complicate encephalomyelitis in mice with delayed appearance of CNS IgM after intranasal alphavirus infection. J Gen Virol 2018 [View Article][PubMed]
    [Google Scholar]
  35. Nakaya T, Sato M, Hata N, Asagiri M, Suemori H et al. Gene induction pathways mediated by distinct IRFs during viral infection. Biochem Biophys Res Commun 2001; 283:1150–1156 [View Article][PubMed]
    [Google Scholar]
  36. Nguyen KB, Watford WT, Salomon R, Hofmann SR, Pien GC et al. Critical role for STAT4 activation by type 1 interferons in the interferon-gamma response to viral infection. Science 2002; 297:2063–2066 [View Article][PubMed]
    [Google Scholar]
  37. der SD, Zhou A, Williams BR, Silverman RH. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci USA 1998; 95:15623–15628 [View Article][PubMed]
    [Google Scholar]
  38. Bréhin AC, Casadémont I, Frenkiel MP, Julier C, Sakuntabhai A et al. The large form of human 2',5'-Oligoadenylate Synthetase (OAS3) exerts antiviral effect against Chikungunya virus. Virology 2009; 384:216–222 [View Article][PubMed]
    [Google Scholar]
  39. Lenschow DJ, Giannakopoulos NV, Gunn LJ, Johnston C, O'Guin AK et al. Identification of interferon-stimulated gene 15 as an antiviral molecule during Sindbis virus infection in vivo. J Virol 2005; 79:13974–13983 [View Article][PubMed]
    [Google Scholar]
  40. Guo X, Ma J, Sun J, Gao G. The zinc-finger antiviral protein recruits the RNA processing exosome to degrade the target mRNA. Proc Natl Acad Sci USA 2007; 104:151–156 [View Article][PubMed]
    [Google Scholar]
  41. Zhang Y, Burke CW, Ryman KD, Klimstra WB. Identification and characterization of interferon-induced proteins that inhibit alphavirus replication. J Virol 2007; 81:11246–11255 [View Article][PubMed]
    [Google Scholar]
  42. Barry G, Breakwell L, Fragkoudis R, Attarzadeh-Yazdi G, Rodriguez-Andres J et al. PKR acts early in infection to suppress Semliki Forest virus production and strongly enhances the type I interferon response. J Gen Virol 2009; 90:1382–1391 [View Article][PubMed]
    [Google Scholar]
  43. Bosco A, Wiehler S, Proud D. Interferon regulatory factor 7 regulates airway epithelial cell responses to human rhinovirus infection. BMC Genomics 2016; 17:76 [View Article][PubMed]
    [Google Scholar]
  44. Semple BD, Kossmann T, Morganti-Kossmann MC. Role of chemokines in CNS health and pathology: a focus on the CCL2/CCR2 and CXCL8/CXCR2 networks. J Cereb Blood Flow Metab 2010; 30:459–473 [View Article][PubMed]
    [Google Scholar]
  45. Baxter VK, Glowinski R, Braxton AM, Potter MC, Slusher BS et al. Glutamine antagonist-mediated immune suppression decreases pathology but delays virus clearance in mice during nonfatal alphavirus encephalomyelitis. Virology 2017; 508:134–149 [View Article][PubMed]
    [Google Scholar]
  46. Rowell JF, Griffin DE. The inflammatory response to nonfatal Sindbis virus infection of the nervous system is more severe in SJL than in BALB/c mice and is associated with low levels of IL-4 mRNA and high levels of IL-10-producing CD4+ T cells. J Immunol 1999; 162:1624–1632[PubMed]
    [Google Scholar]
  47. Marsili G, Perrotti E, Remoli AL, Acchioni C, Sgarbanti M et al. IFN regulatory factors and antiviral innate immunity: how viruses can get better. J Interferon Cytokine Res 2016; 36:414–432 [View Article][PubMed]
    [Google Scholar]
  48. Honda K, Takaoka A, Taniguchi T. Type I interferon [corrected] gene induction by the interferon regulatory factor family of transcription factors. Immunity 2006; 25:349–360 [View Article][PubMed]
    [Google Scholar]
  49. Ousman SS, Wang J, Campbell IL. Differential regulation of interferon regulatory factor (IRF)-7 and IRF-9 gene expression in the central nervous system during viral infection. J Virol 2005; 79:7514–7527 [View Article][PubMed]
    [Google Scholar]
  50. Delhaye S, Paul S, Blakqori G, Minet M, Weber F et al. Neurons produce type I interferon during viral encephalitis. Proc Natl Acad Sci USA 2006; 103:7835–7840 [View Article][PubMed]
    [Google Scholar]
  51. Cohen M, Matcovitch O, David E, Barnett-Itzhaki Z, Keren-Shaul H et al. Chronic exposure to TGFβ1 regulates myeloid cell inflammatory response in an IRF7-dependent manner. EMBO J 2014; 33:2906–2921 [View Article][PubMed]
    [Google Scholar]
  52. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ et al. Identification of a unique TGF-β-dependent molecular and functional signature in microglia. Nat Neurosci 2014; 17:131–143 [View Article][PubMed]
    [Google Scholar]
  53. Hagemeyer N, Prinz M. Burning down the house: IRF7 makes the difference for microglia. EMBO J 2014; 33:2885–2886 [View Article][PubMed]
    [Google Scholar]
  54. Juang YT, Lowther W, Kellum M, Au WC, Lin R et al. Primary activation of interferon A and interferon B gene transcription by interferon regulatory factor 3. Proc Natl Acad Sci USA 1998; 95:9837–9842 [View Article][PubMed]
    [Google Scholar]
  55. Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E et al. IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol 2003; 4:491–496 [View Article][PubMed]
    [Google Scholar]
  56. Sharma S, Tenoever BR, Grandvaux N, Zhou GP, Lin R et al. Triggering the interferon antiviral response through an IKK-related pathway. Science 2003; 300:1148–1151 [View Article][PubMed]
    [Google Scholar]
  57. Servant MJ, Grandvaux N, Hiscott J. Multiple signaling pathways leading to the activation of interferon regulatory factor 3. Biochem Pharmacol 2002; 64:985–992 [View Article][PubMed]
    [Google Scholar]
  58. Doyle S, Vaidya S, O'Connell R, Dadgostar H, Dempsey P et al. IRF3 mediates a TLR3/TLR4-specific antiviral gene program. Immunity 2002; 17:251–263 [View Article][PubMed]
    [Google Scholar]
  59. Lazear HM, Govero J, Smith AM, Platt DJ, Fernandez E et al. A mouse model of Zika virus pathogenesis. Cell Host Microbe 2016; 19:720–730 [View Article][PubMed]
    [Google Scholar]
  60. Chen HW, King K, Tu J, Sanchez M, Luster AD et al. The roles of IRF-3 and IRF-7 in innate antiviral immunity against dengue virus. J Immunol 2013; 191:4194–4201 [View Article][PubMed]
    [Google Scholar]
  61. Schilte C, Buckwalter MR, Laird ME, Diamond MS, Schwartz O et al. Cutting edge: independent roles for IRF-3 and IRF-7 in hematopoietic and nonhematopoietic cells during host response to Chikungunya infection. J Immunol 2012; 188:2967–2971 [View Article][PubMed]
    [Google Scholar]
  62. Daffis S, Lazear HM, Liu WJ, Audsley M, Engle M et al. The naturally attenuated Kunjin strain of West Nile virus shows enhanced sensitivity to the host type I interferon response. J Virol 2011; 85:5664–5668 [View Article][PubMed]
    [Google Scholar]
  63. Thackray LB, Duan E, Lazear HM, Kambal A, Schreiber RD et al. Critical role for interferon regulatory factor 3 (IRF-3) and IRF-7 in type I interferon-mediated control of murine norovirus replication. J Virol 2012; 86:13515–13523 [View Article][PubMed]
    [Google Scholar]
  64. Proenca-Modena JL, Sesti-Costa R, Pinto AK, Richner JM, Lazear HM et al. Oropouche virus infection and pathogenesis are restricted by MAVS, IRF-3, IRF-7, and type I interferon signaling pathways in nonmyeloid cells. J Virol 2015; 89:4720–4737 [View Article][PubMed]
    [Google Scholar]
  65. Chattopadhyay S, Kuzmanovic T, Zhang Y, Wetzel JL, Sen GC. Ubiquitination of the transcription factor IRF-3 activates RIPA, the apoptotic pathway that protects mice from viral pathogenesis. Immunity 2016; 44:1151–1161 [View Article][PubMed]
    [Google Scholar]
  66. Moore TC, Vogel AJ, Petro TM, Brown DM. IRF3 deficiency impacts granzyme B expression and maintenance of memory T cell function in response to viral infection. Microbes Infect 2015; 17:426–439 [View Article][PubMed]
    [Google Scholar]
  67. Tarassishin L, Suh HS, Lee SC. Interferon regulatory factor 3 plays an anti-inflammatory role in microglia by activating the PI3K/Akt pathway. J Neuroinflammation 2011; 8:187 [View Article][PubMed]
    [Google Scholar]
  68. Chistiakov DA, Myasoedova VA, Revin VV, Orekhov AN, Bobryshev YV. The impact of interferon-regulatory factors to macrophage differentiation and polarization into M1 and M2. Immunobiology 2018; 223:101–111 [View Article][PubMed]
    [Google Scholar]
  69. Ysebrant de Lendonck L, Martinet V, Goriely S. Interferon regulatory factor 3 in adaptive immune responses. Cell Mol Life Sci 2014; 71:3873–3883 [View Article][PubMed]
    [Google Scholar]
  70. Fitzgerald DC, O'Brien K, Young A, Fonseca-Kelly Z, Rostami A et al. Interferon regulatory factor (IRF) 3 is critical for the development of experimental autoimmune encephalomyelitis. J Neuroinflammation 2014; 11:130 [View Article][PubMed]
    [Google Scholar]
  71. Kulcsar KA, Baxter VK, Greene IP, Griffin DE. Interleukin 10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyelitis. Proc Natl Acad Sci USA 2014; 111:16053–16058 [View Article][PubMed]
    [Google Scholar]
  72. Chattopadhyay S, Marques JT, Yamashita M, Peters KL, Smith K et al. Viral apoptosis is induced by IRF-3-mediated activation of Bax. EMBO J 2010; 29:1762–1773 [View Article][PubMed]
    [Google Scholar]
  73. Chattopadhyay S, Fensterl V, Zhang Y, Veleeparambil M, Yamashita M et al. Role of interferon regulatory factor 3-mediated apoptosis in the establishment and maintenance of persistent infection by Sendai virus. J Virol 2013; 87:16–24 [View Article][PubMed]
    [Google Scholar]
  74. Chattopadhyay S, Yamashita M, Zhang Y, Sen GC. The IRF-3/Bax-mediated apoptotic pathway, activated by viral cytoplasmic RNA and DNA, inhibits virus replication. J Virol 2011; 85:3708–3716 [View Article][PubMed]
    [Google Scholar]
  75. Peters K, Chattopadhyay S, Sen GC. IRF-3 activation by Sendai virus infection is required for cellular apoptosis and avoidance of persistence. J Virol 2008; 82:3500–3508 [View Article][PubMed]
    [Google Scholar]
  76. Peltier DC, Lazear HM, Farmer JR, Diamond MS, Miller DJ. Neurotropic arboviruses induce interferon regulatory factor 3-mediated neuronal responses that are cytoprotective, interferon independent, and inhibited by Western equine encephalitis virus capsid. J Virol 2013; 87:1821–1833 [View Article][PubMed]
    [Google Scholar]
  77. Zhou S, Cerny AM, Fitzgerald KA, Kurt-Jones EA, Finberg RW. Role of interferon regulatory factor 7 in T cell responses during acute lymphocytic choriomeningitis virus infection. J Virol 2012; 86:11254–11265 [View Article][PubMed]
    [Google Scholar]
  78. Barnes BJ, Richards J, Mancl M, Hanash S, Beretta L et al. Global and distinct targets of IRF-5 and IRF-7 during innate response to viral infection. J Biol Chem 2004; 279:45194–45207 [View Article][PubMed]
    [Google Scholar]
  79. Jiang DS, Liu Y, Zhou H, Zhang Y, Zhang XD et al. Interferon regulatory factor 7 functions as a novel negative regulator of pathological cardiac hypertrophy. Hypertension 2014; 63:713–722 [View Article][PubMed]
    [Google Scholar]
  80. Rollenhagen C, Rollenhage C, Macura SL, Lathrop MJ, Mackenzie TA et al. Enhancing interferon regulatory factor 7 mediated antiviral responses and decreasing nuclear factor κB expression limit HIV-1 replication in cervical tissues. PLoS One 2015; 10:e0131919 [View Article][PubMed]
    [Google Scholar]
  81. Freter RR, Alberta JA, Hwang GY, Wrentmore AL, Stiles CD. Platelet-derived growth factor induction of the immediate-early gene MCP-1 is mediated by NF-κB and a 90-kDa phosphoprotein coactivator. J Biol Chem 1996; 271:17417–17424 [View Article][PubMed]
    [Google Scholar]
  82. Spann KM, Loh Z, Lynch JP, Ullah A, Zhang V et al. IRF-3, IRF-7, and IPS-1 promote host defense against acute human metapneumovirus infection in neonatal mice. Am J Pathol 2014; 184:1795–1806 [View Article][PubMed]
    [Google Scholar]
  83. Li W, Hofer MJ, Jung SR, Lim SL, Campbell IL. IRF7-dependent type I interferon production induces lethal immune-mediated disease in STAT1 knockout mice infected with lymphocytic choriomeningitis virus. J Virol 2014; 88:7578–7588 [View Article][PubMed]
    [Google Scholar]
  84. Christensen JE, Fenger C, Issazadeh-Navikas S, Krug A, Liljestrøm P et al. Differential impact of interferon regulatory factor 7 in initiation of the type I interferon response in the lymphocytic choriomeningitis virus-infected central nervous system versus the periphery. J Virol 2012; 86:7384–7392 [View Article][PubMed]
    [Google Scholar]
  85. Nargi-Aizenman JL, Havert MB, Zhang M, Irani DN, Rothstein JD et al. Glutamate receptor antagonists protect from virus-induced neural degeneration. Ann Neurol 2004; 55:541–549 [View Article][PubMed]
    [Google Scholar]
  86. Manivannan S, Baxter VK, Schultz KL, Slusher BS, Griffin DE. Protective effects of glutamine antagonist 6-diazo-5-oxo-l-norleucine in mice with alphavirus encephalomyelitis. J Virol 2016; 90:9251–9262 [View Article][PubMed]
    [Google Scholar]
  87. Greene IP, Lee EY, Prow N, Ngwang B, Griffin DE. Protection from fatal viral encephalomyelitis: AMPA receptor antagonists have a direct effect on the inflammatory response to infection. Proc Natl Acad Sci USA 2008; 105:3575–3580 [View Article][PubMed]
    [Google Scholar]
  88. Schultz KL, Vernon PS, Griffin DE. Differentiation of neurons restricts Arbovirus replication and increases expression of the alpha isoform of IRF-7. J Virol 2015; 89:48–60 [View Article][PubMed]
    [Google Scholar]
  89. Xu D, Zhang Y, Zhao L, Cao M, Lingel A et al. Interferon regulatory factor 7 is involved in the growth of Epstein-Barr virus-transformed human B lymphocytes. Virus Res 2015; 195:112–118 [View Article][PubMed]
    [Google Scholar]
  90. Carmen J, Rothstein JD, Kerr DA. Tumor necrosis factor-alpha modulates glutamate transport in the CNS and is a critical determinant of outcome from viral encephalomyelitis. Brain Res 2009; 1263:143–154 [View Article][PubMed]
    [Google Scholar]
  91. Kraft AD, McPherson CA, Harry GJ. Heterogeneity of microglia and TNF signaling as determinants for neuronal death or survival. Neurotoxicology 2009; 30:785–793 [View Article][PubMed]
    [Google Scholar]
  92. Allan SM, Rothwell NJ. Cytokines and acute neurodegeneration. Nat Rev Neurosci 2001; 2:734–744 [View Article][PubMed]
    [Google Scholar]
  93. Park KM, Bowers WJ. Tumor necrosis factor-alpha mediated signaling in neuronal homeostasis and dysfunction. Cell Signal 2010; 22:977–983 [View Article][PubMed]
    [Google Scholar]
  94. Yu Z, Cheng G, Wen X, Wu GD, Lee WT et al. Tumor necrosis factor alpha increases neuronal vulnerability to excitotoxic necrosis by inducing expression of the AMPA-glutamate receptor subunit GluR1 via an acid sphingomyelinase- and NFκB-dependent mechanism. Neurobiol Dis 2002; 11:199–213 [View Article][PubMed]
    [Google Scholar]
  95. Ferguson AR, Christensen RN, Gensel JC, Miller BA, Sun F et al. Cell death after spinal cord injury is exacerbated by rapid TNF α-induced trafficking of GluR2-lacking AMPARs to the plasma membrane. J Neurosci 2008; 28:11391–11400 [View Article][PubMed]
    [Google Scholar]
  96. Venters HD, Dantzer R, Kelley KW. A new concept in neurodegeneration: TNFalpha is a silencer of survival signals. Trends Neurosci 2000; 23:175–180 [View Article][PubMed]
    [Google Scholar]
  97. Tolosa L, Caraballo-Miralles V, Olmos G, Lladó J. TNF-α potentiates glutamate-induced spinal cord motoneuron death via NF-κB. Mol Cell Neurosci 2011; 46:176–186 [View Article][PubMed]
    [Google Scholar]
  98. Brenner D, Blaser H, Mak TW. Regulation of tumour necrosis factor signalling: live or let die. Nat Rev Immunol 2015; 15:362–374 [View Article][PubMed]
    [Google Scholar]
  99. Tortarolo M, Vallarola A, Lidonnici D, Battaglia E, Gensano F et al. Lack of TNF-α receptor type 2 protects motor neurons in a cellular model of amyotrophic lateral sclerosis and in mutant SOD1 mice but does not affect disease progression. J Neurochem 2015; 135:109–124 [View Article][PubMed]
    [Google Scholar]
  100. Toribio R, Ventoso I. Inhibition of host translation by virus infection in vivo. Proc Natl Acad Sci USA 2010; 107:9837–9842 [View Article][PubMed]
    [Google Scholar]
  101. Wang L, Du F, Wang X. TNF-alpha induces two distinct caspase-8 activation pathways. Cell 2008; 133:693–703 [View Article][PubMed]
    [Google Scholar]
  102. Chang A, Chen Y, Shen W, Gao R, Zhou W et al. Ifit1 protects against lipopolysaccharide and d-galactosamine-induced fatal hepatitis by inhibiting activation of the JNK pathway. J Infect Dis 2015; 212:1509–1520 [View Article][PubMed]
    [Google Scholar]
  103. John SP, Sun J, Carlson RJ, Cao B, Bradfield CJ et al. IFIT1 exerts opposing regulatory effects on the inflammatory and interferon gene programs in LPS-activated human macrophages. Cell Rep 2018; 25:95–106 [View Article][PubMed]
    [Google Scholar]
  104. Zhang ZJ, Jiang BC, Gao YJ. Chemokines in neuron-glial cell interaction and pathogenesis of neuropathic pain. Cell Mol Life Sci 2017; 74:3275–3291 [View Article][PubMed]
    [Google Scholar]
  105. Karrer M, Lopez MA, Meier D, Mikhail C, Ogunshola OO et al. Cytokine-induced sleep: neurons respond to TNF with production of chemokines and increased expression of Homer1a in vitro. Brain Behav Immun 2015; 47:186–192 [View Article][PubMed]
    [Google Scholar]
  106. Bennett JL, Elhofy A, Canto MC, Tani M, Ransohoff RM et al. CCL2 transgene expression in the central nervous system directs diffuse infiltration of CD45(high)CD11b(+) monocytes and enhanced Theiler's murine encephalomyelitis virus-induced demyelinating disease. J Neurovirol 2003; 9:623–636[PubMed]
    [Google Scholar]
  107. Terry RL, Getts DR, Deffrasnes C, van Vreden C, Campbell IL et al. Inflammatory monocytes and the pathogenesis of viral encephalitis. J Neuroinflammation 2012; 9:270 [View Article][PubMed]
    [Google Scholar]
  108. Getts DR, Terry RL, Getts MT, Müller M, Rana S et al. Ly6c+ "inflammatory monocytes" are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J Exp Med 2008; 205:2319–2337 [View Article][PubMed]
    [Google Scholar]
  109. Lane TE, Asensio VC, Yu N, Paoletti AD, Campbell IL et al. Dynamic regulation of alpha- and β-chemokine expression in the central nervous system during mouse hepatitis virus-induced demyelinating disease. J Immunol 1998; 160:970–978[PubMed]
    [Google Scholar]
  110. Templeton SP, Kim TS, O'Malley K, Perlman S. Maturation and localization of macrophages and microglia during infection with a neurotropic murine coronavirus. Brain Pathol 2008; 18:40–51 [View Article][PubMed]
    [Google Scholar]
  111. Trujillo JA, Fleming EL, Perlman S. Transgenic CCL2 expression in the central nervous system results in a dysregulated immune response and enhanced lethality after coronavirus infection. J Virol 2013; 87:2376–2389 [View Article][PubMed]
    [Google Scholar]
  112. Luther SA, Ansel KM, Cyster JG. Overlapping roles of CXCL13, interleukin 7 receptor alpha, and CCR7 ligands in lymph node development. J Exp Med 2003; 197:1191–1198 [View Article][PubMed]
    [Google Scholar]
  113. Shi K, Hayashida K, Kaneko M, Hashimoto J, Tomita T et al. Lymphoid chemokine B cell-attracting chemokine-1 (CXCL13) is expressed in germinal center of ectopic lymphoid follicles within the synovium of chronic arthritis patients. J Immunol 2001; 166:650–655 [View Article][PubMed]
    [Google Scholar]
  114. Bagaeva LV, Rao P, Powers JM, Segal BM. CXC chemokine ligand 13 plays a role in experimental autoimmune encephalomyelitis. J Immunol 2006; 176:7676–7685 [View Article][PubMed]
    [Google Scholar]
  115. Aloisi F, Columba-Cabezas S, Franciotta D, Rosicarelli B, Magliozzi R et al. Lymphoid chemokines in chronic neuroinflammation. J Neuroimmunol 2008; 198:106–112 [View Article][PubMed]
    [Google Scholar]
  116. Kobayashi S, Watanabe T, Suzuki R, Furu M, Ito H et al. TGF-β induces the differentiation of human CXCL13-producing CD4(+) T cells. Eur J Immunol 2016; 46:360–371 [View Article][PubMed]
    [Google Scholar]
  117. Rainey-Barger EK, Rumble JM, Lalor SJ, Esen N, Segal BM et al. The lymphoid chemokine, CXCL13, is dispensable for the initial recruitment of B cells to the acutely inflamed central nervous system. Brain Behav Immun 2011; 25:922–931 [View Article][PubMed]
    [Google Scholar]
  118. Metcalf TU, Baxter VK, Nilaratanakul V, Griffin DE. Recruitment and retention of B cells in the central nervous system in response to alphavirus encephalomyelitis. J Virol 2013; 87:2420–2429 [View Article][PubMed]
    [Google Scholar]
  119. Phares TW, Disano KD, Stohlman SA, Segal BM, Bergmann CC. CXCL13 promotes isotype-switched B cell accumulation to the central nervous system during viral encephalomyelitis. Brain Behav Immun 2016; 54:128–139 [View Article][PubMed]
    [Google Scholar]
  120. Esen N, Blakely PK, Rainey-Barger EK, Irani DN. Complexity of the microglial activation pathways that drive innate host responses during lethal alphavirus encephalitis in mice. ASN Neuro 2012; 4:207–221 [View Article][PubMed]
    [Google Scholar]
  121. Müller U, Steinhoff U, Reis LF, Hemmi S, Pavlovic J et al. Functional role of type I and type II interferons in antiviral defense. Science 1994; 264:1918–1921 [View Article][PubMed]
    [Google Scholar]
  122. Lustig S, Jackson AC, Hahn CS, Griffin DE, Strauss EG et al. Molecular basis of Sindbis virus neurovirulence in mice. J Virol 1988; 62:2329–2336[PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001174
Loading
/content/journal/jgv/10.1099/jgv.0.001174
Loading

Data & Media loading...

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