Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Tick-borne encephalitis virus induces chemokine RANTES expression via activation of IRF-3 pathway

Authors: Xiaowei Zhang, Zhenhua Zheng, Xijuan Liu, Bo Shu, Panyong Mao, Bingke Bai, Qinxue Hu, Minhua Luo, Xiaohe Ma, Zongqiang Cui, Hanzhong Wang

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Tick-borne encephalitis virus (TBEV) is one of the most important flaviviruses that targets the central nervous system (CNS) and causes encephalitides in humans. Although neuroinflammatory mechanisms may contribute to brain tissue destruction, the induction pathways and potential roles of specific chemokines in TBEV-mediated neurological disease are poorly understood.

Methods

BALB/c mice were intracerebrally injected with TBEV, followed by evaluation of chemokine and cytokine profiles using protein array analysis. The virus-infected mice were treated with the CC chemokine antagonist Met-RANTES or anti-RANTES mAb to determine the role of RANTES in affecting TBEV-induced neurological disease. The underlying signaling mechanisms were delineated using RANTES promoter luciferase reporter assay, siRNA-mediated knockdown, and pharmacological inhibitors in human brain-derived cell culture models.

Results

In a mouse model, pathological features including marked inflammatory cell infiltrates were observed in brain sections, which correlated with a robust up-regulation of RANTES within the brain but not in peripheral tissues and sera. Antagonizing RANTES within CNS extended the survival of mice and reduced accumulation of infiltrating cells in the brain after TBEV infection. Through in vitro studies, we show that virus infection up-regulated RANTES production at both mRNA and protein levels in human brain-derived cell lines and primary progenitor-derived astrocytes. Furthermore, IRF-3 pathway appeared to be essential for TBEV-induced RANTES production. Site mutation of an IRF-3-binding motif abrogated the RANTES promoter activity in virus-infected brain cells. Moreover, IRF-3 was activated upon TBEV infection as evidenced by phosphorylation of TBK1 and IRF-3, while blockade of IRF-3 activation drastically reduced virus-induced RANTES expression.

Conclusions

Our findings together provide insights into the molecular mechanism underlying RANTES production induced by TBEV, highlighting its potential importance in the process of neuroinflammatory responses to TBEV infection.
Appendix
Available only for authorised users
Literature
2.
go back to reference Mansfield KL, Johnson N, Phipps LP, Stephenson JR, Fooks AR, Solomon T. Tick-borne encephalitis virus—a review of an emerging zoonosis. J Gen Virol. 2009;90(8):1781–94.CrossRefPubMed Mansfield KL, Johnson N, Phipps LP, Stephenson JR, Fooks AR, Solomon T. Tick-borne encephalitis virus—a review of an emerging zoonosis. J Gen Virol. 2009;90(8):1781–94.CrossRefPubMed
4.
go back to reference Gritsun T, Frolova T, Zhankov A, Armesto M, Turner S, Frolova M, et al. Characterization of a siberian virus isolated from a patient with progressive chronic tick-borne encephalitis. J Virol. 2003;77(1):25–36.CrossRefPubMedPubMedCentral Gritsun T, Frolova T, Zhankov A, Armesto M, Turner S, Frolova M, et al. Characterization of a siberian virus isolated from a patient with progressive chronic tick-borne encephalitis. J Virol. 2003;77(1):25–36.CrossRefPubMedPubMedCentral
5.
go back to reference Mandl CW. Steps of the tick-borne encephalitis virus replication cycle that affect neuropathogenesis. Virus Res. 2005;111(2):161–74.CrossRefPubMed Mandl CW. Steps of the tick-borne encephalitis virus replication cycle that affect neuropathogenesis. Virus Res. 2005;111(2):161–74.CrossRefPubMed
6.
go back to reference Bogovic P, Strle F. Tick-borne encephalitis: a review of epidemiology, clinical characteristics, and management. World J Clin Cases. 2015;3(5):430–41.CrossRefPubMedPubMedCentral Bogovic P, Strle F. Tick-borne encephalitis: a review of epidemiology, clinical characteristics, and management. World J Clin Cases. 2015;3(5):430–41.CrossRefPubMedPubMedCentral
7.
go back to reference Gelpi E, Preusser M, Garzuly F, Holzmann H, Heinz FX, Budka H. Visualization of Central European tick-borne encephalitis infection in fatal human cases. J Neuropathol Exp Neurol. 2005;64(6):506–12.CrossRefPubMed Gelpi E, Preusser M, Garzuly F, Holzmann H, Heinz FX, Budka H. Visualization of Central European tick-borne encephalitis infection in fatal human cases. J Neuropathol Exp Neurol. 2005;64(6):506–12.CrossRefPubMed
8.
go back to reference Gelpi E, Preusser M, Laggner U, Garzuly F, Holzmann H, Heinz FX, et al. Inflammatory response in human tick-borne encephalitis: analysis of postmortem brain tissue. J Neurovirol. 2006;12(4):322–7.CrossRefPubMed Gelpi E, Preusser M, Laggner U, Garzuly F, Holzmann H, Heinz FX, et al. Inflammatory response in human tick-borne encephalitis: analysis of postmortem brain tissue. J Neurovirol. 2006;12(4):322–7.CrossRefPubMed
9.
go back to reference Růžek D, Salát J, Palus M, Gritsun TS, Gould EA, Dyková I, et al. CD8+ T-cells mediate immunopathology in tick-borne encephalitis. Virology. 2009;384(1):1–6.CrossRefPubMed Růžek D, Salát J, Palus M, Gritsun TS, Gould EA, Dyková I, et al. CD8+ T-cells mediate immunopathology in tick-borne encephalitis. Virology. 2009;384(1):1–6.CrossRefPubMed
10.
go back to reference Lepej SZ, Misic-Majerus L, Jeren T, Rode OD, Remenar A, Sporec V, et al. Chemokines CXCL10 and CXCL11 in the cerebrospinal fluid of patients with tick-borne encephalitis. Acta Neurol Scand. 2007;115(2):109–14.CrossRefPubMed Lepej SZ, Misic-Majerus L, Jeren T, Rode OD, Remenar A, Sporec V, et al. Chemokines CXCL10 and CXCL11 in the cerebrospinal fluid of patients with tick-borne encephalitis. Acta Neurol Scand. 2007;115(2):109–14.CrossRefPubMed
11.
go back to reference Zajkowska J, Moniuszko-Malinowska A, Pancewicz SA, Muszynska-Mazur A, Kondrusik M, Grygorczuk S, et al. Evaluation of CXCL10, CXCL11, CXCL12 and CXCL13 chemokines in serum and cerebrospinal fluid in patients with tick borne encephalitis (TBE). Adv Med Sci. 2011;56(2):311–7.CrossRefPubMed Zajkowska J, Moniuszko-Malinowska A, Pancewicz SA, Muszynska-Mazur A, Kondrusik M, Grygorczuk S, et al. Evaluation of CXCL10, CXCL11, CXCL12 and CXCL13 chemokines in serum and cerebrospinal fluid in patients with tick borne encephalitis (TBE). Adv Med Sci. 2011;56(2):311–7.CrossRefPubMed
12.
go back to reference Grygorczuk S, Zajkowska J, Swierzbinska R, Pancewicz S, Kondrusik M, Hermanowska-Szpakowicz T. Concentration of the beta-chemokine CCL5 (RANTES) in cerebrospinal fluid in patients with tick-borne encephalitis. Neurol Neurochir Pol. 2006;40(2):106–11.PubMed Grygorczuk S, Zajkowska J, Swierzbinska R, Pancewicz S, Kondrusik M, Hermanowska-Szpakowicz T. Concentration of the beta-chemokine CCL5 (RANTES) in cerebrospinal fluid in patients with tick-borne encephalitis. Neurol Neurochir Pol. 2006;40(2):106–11.PubMed
13.
go back to reference Michałowska-Wender G, Losy J, Kondrusik M, Zajkowska J, Pancewicz S, Grygorczuk S, et al. Evaluation of soluble platelet cell adhesion molecule sPECAM-1 and chemokine MCP-1 (CCL2) concentration in CSF of patients with tick-borne encephalitis. Pol Merkur Lekarski. 2006;20(115):46–8.PubMed Michałowska-Wender G, Losy J, Kondrusik M, Zajkowska J, Pancewicz S, Grygorczuk S, et al. Evaluation of soluble platelet cell adhesion molecule sPECAM-1 and chemokine MCP-1 (CCL2) concentration in CSF of patients with tick-borne encephalitis. Pol Merkur Lekarski. 2006;20(115):46–8.PubMed
14.
go back to reference Grygorczuk S, Osada J, Parczewski M, Moniuszko A, Swierzbinska R, Kondrusik M, et al. The expression of the chemokine receptor CCR5 in tick-borne encephalitis. J Neuroinflammation. 2016;13:45.CrossRefPubMedPubMedCentral Grygorczuk S, Osada J, Parczewski M, Moniuszko A, Swierzbinska R, Kondrusik M, et al. The expression of the chemokine receptor CCR5 in tick-borne encephalitis. J Neuroinflammation. 2016;13:45.CrossRefPubMedPubMedCentral
16.
go back to reference Epstein FH, Luster AD. Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med. 1998;338(7):436–45.CrossRef Epstein FH, Luster AD. Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med. 1998;338(7):436–45.CrossRef
18.
go back to reference Zhang X, Zheng Z, Shu B, Mao P, Bai B, Hu Q, et al. Isolation and characterization of a Far-Eastern strain of tick-borne encephalitis virus in China. Virus Res. 2016;213:6–10.CrossRefPubMed Zhang X, Zheng Z, Shu B, Mao P, Bai B, Hu Q, et al. Isolation and characterization of a Far-Eastern strain of tick-borne encephalitis virus in China. Virus Res. 2016;213:6–10.CrossRefPubMed
19.
go back to reference Luo MH, Schwartz PH, Fortunato EA. Neonatal neural progenitor cells and their neuronal and glial cell derivatives are fully permissive for human cytomegalovirus infection. J Virol. 2008;82(20):9994–10007.CrossRefPubMedPubMedCentral Luo MH, Schwartz PH, Fortunato EA. Neonatal neural progenitor cells and their neuronal and glial cell derivatives are fully permissive for human cytomegalovirus infection. J Virol. 2008;82(20):9994–10007.CrossRefPubMedPubMedCentral
20.
go back to reference Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, et al. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013;87(22):12407–21.CrossRefPubMedPubMedCentral Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, et al. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013;87(22):12407–21.CrossRefPubMedPubMedCentral
21.
go back to reference Daigneault M, Preston JA, Marriott HM, Whyte MKB, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS ONE. 2010;5(1):e8668.CrossRefPubMedPubMedCentral Daigneault M, Preston JA, Marriott HM, Whyte MKB, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS ONE. 2010;5(1):e8668.CrossRefPubMedPubMedCentral
22.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001;25(4):402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001;25(4):402–8.CrossRefPubMed
23.
go back to reference Yen Y-T, Liao F, Hsiao C-H, Kao C-L, Chen Y-C, Wu-Hsieh BA. Modeling the early events of severe acute respiratory syndrome coronavirus infection in vitro. J Virol. 2006;80(6):2684–93.CrossRefPubMedPubMedCentral Yen Y-T, Liao F, Hsiao C-H, Kao C-L, Chen Y-C, Wu-Hsieh BA. Modeling the early events of severe acute respiratory syndrome coronavirus infection in vitro. J Virol. 2006;80(6):2684–93.CrossRefPubMedPubMedCentral
24.
go back to reference Veldkamp CT, Seibert C, Peterson FC, De la Cruz NB, Haugner III JC, Basnet H, et al. Structural basis of CXCR4 sulfotyrosine recognition by the chemokine SDF-1/CXCL12. Sci Signal. 2008;1(37):ra4.CrossRefPubMedPubMedCentral Veldkamp CT, Seibert C, Peterson FC, De la Cruz NB, Haugner III JC, Basnet H, et al. Structural basis of CXCR4 sulfotyrosine recognition by the chemokine SDF-1/CXCL12. Sci Signal. 2008;1(37):ra4.CrossRefPubMedPubMedCentral
25.
go back to reference Marino APM, da Silva A, dos Santos P, de Oliveira Pinto LM, Gazzinelli RT, Teixeira MM, et al. Regulated on activation, normal T cell expressed and secreted (RANTES) antagonist (Met-RANTES) controls the early phase of Trypanosoma cruzi-elicited myocarditis. Circulation. 2004;110(11):1443–9.CrossRefPubMed Marino APM, da Silva A, dos Santos P, de Oliveira Pinto LM, Gazzinelli RT, Teixeira MM, et al. Regulated on activation, normal T cell expressed and secreted (RANTES) antagonist (Met-RANTES) controls the early phase of Trypanosoma cruzi-elicited myocarditis. Circulation. 2004;110(11):1443–9.CrossRefPubMed
26.
go back to reference Proudfoot AE, Power CA, Hoogewerf AJ, Montjovent MO, Borlat F, Offord RE, et al. Extension of recombinant human RANTES by the retention of the initiating methionine produces a potent antagonist. J Biol Chem. 1996;271(5):2599–603.CrossRefPubMed Proudfoot AE, Power CA, Hoogewerf AJ, Montjovent MO, Borlat F, Offord RE, et al. Extension of recombinant human RANTES by the retention of the initiating methionine produces a potent antagonist. J Biol Chem. 1996;271(5):2599–603.CrossRefPubMed
27.
go back to reference Di Liberto D, Locati M, Caccamo N, Vecchi A, Meraviglia S, Salerno A, et al. Role of the chemokine decoy receptor D6 in balancing inflammation, immune activation, and antimicrobial resistance in Mycobacterium tuberculosis infection. J Exp Med. 2008;205(9):2075–84.CrossRefPubMedPubMedCentral Di Liberto D, Locati M, Caccamo N, Vecchi A, Meraviglia S, Salerno A, et al. Role of the chemokine decoy receptor D6 in balancing inflammation, immune activation, and antimicrobial resistance in Mycobacterium tuberculosis infection. J Exp Med. 2008;205(9):2075–84.CrossRefPubMedPubMedCentral
28.
go back to reference Cruickshank SM, Deschoolmeester ML, Svensson M, Howell G, Bazakou A, Logunova L, et al. Rapid dendritic cell mobilization to the large intestinal epithelium is associated with resistance to Trichuris muris infection. J Immunol. 2009;182(5):3055–62.CrossRefPubMedPubMedCentral Cruickshank SM, Deschoolmeester ML, Svensson M, Howell G, Bazakou A, Logunova L, et al. Rapid dendritic cell mobilization to the large intestinal epithelium is associated with resistance to Trichuris muris infection. J Immunol. 2009;182(5):3055–62.CrossRefPubMedPubMedCentral
29.
go back to reference Casola A, Garofalo RP, Haeberle H, Elliott TF, Lin R, Jamaluddin M, et al. Multiple cis regulatory elements control RANTES promoter activity in alveolar epithelial cells infected with respiratory syncytial virus. J Virol. 2001;75(14):6428–39.CrossRefPubMedPubMedCentral Casola A, Garofalo RP, Haeberle H, Elliott TF, Lin R, Jamaluddin M, et al. Multiple cis regulatory elements control RANTES promoter activity in alveolar epithelial cells infected with respiratory syncytial virus. J Virol. 2001;75(14):6428–39.CrossRefPubMedPubMedCentral
30.
go back to reference Zhang P, Li Y, Xia J, He J, Pu J, Xie J, et al. IPS-1 plays an essential role in dsRNA-induced stress granule formation by interacting with PKR and promoting its activation. J Cell Sci. 2014;127(Pt 11):2471–82.CrossRefPubMed Zhang P, Li Y, Xia J, He J, Pu J, Xie J, et al. IPS-1 plays an essential role in dsRNA-induced stress granule formation by interacting with PKR and promoting its activation. J Cell Sci. 2014;127(Pt 11):2471–82.CrossRefPubMed
31.
go back to reference Clark K, Plater L, Peggie M, Cohen P. Use of the pharmacological inhibitor BX795 to study the regulation and physiological roles of TBK1 and IkB kinase ε: a distinct upstream kinase mediates Ser-172 phosphorylation and activation. J Biol Chem. 2009;284(21):14136–46.CrossRefPubMedPubMedCentral Clark K, Plater L, Peggie M, Cohen P. Use of the pharmacological inhibitor BX795 to study the regulation and physiological roles of TBK1 and IkB kinase ε: a distinct upstream kinase mediates Ser-172 phosphorylation and activation. J Biol Chem. 2009;284(21):14136–46.CrossRefPubMedPubMedCentral
32.
go back to reference La Ferla-Brühl K, Westhoff M, Karl S, Kasperczyk H, Zwacka R, Debatin K, et al. NF-kB-independent sensitization of glioblastoma cells for TRAIL-induced apoptosis by proteasome inhibition. Oncogene. 2007;26(4):571–82.CrossRefPubMed La Ferla-Brühl K, Westhoff M, Karl S, Kasperczyk H, Zwacka R, Debatin K, et al. NF-kB-independent sensitization of glioblastoma cells for TRAIL-induced apoptosis by proteasome inhibition. Oncogene. 2007;26(4):571–82.CrossRefPubMed
34.
go back to reference McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC, Golenbock DT, Maniatis T. IFN-regulatory factor 3-dependent gene expression is defective in Tbk1-deficient mouse embryonic fibroblasts. Proc Natl Acad Sci U S A. 2004;101(1):233–8.CrossRefPubMed McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC, Golenbock DT, Maniatis T. IFN-regulatory factor 3-dependent gene expression is defective in Tbk1-deficient mouse embryonic fibroblasts. Proc Natl Acad Sci U S A. 2004;101(1):233–8.CrossRefPubMed
35.
go back to reference King NJ, Getts DR, Getts MT, Rana S, Shrestha B, Kesson AM. Immunopathology of flavivirus infections. Immunol Cell Biol. 2006;85(1):33–42.CrossRefPubMed King NJ, Getts DR, Getts MT, Rana S, Shrestha B, Kesson AM. Immunopathology of flavivirus infections. Immunol Cell Biol. 2006;85(1):33–42.CrossRefPubMed
36.
go back to reference Bily T, Palus M, Eyer L, Elsterova J, Vancova M, Ruzek D. Electron tomography analysis of tick-borne encephalitis virus infection in human neurons. Sci Rep. 2015;5:10745.CrossRefPubMedPubMedCentral Bily T, Palus M, Eyer L, Elsterova J, Vancova M, Ruzek D. Electron tomography analysis of tick-borne encephalitis virus infection in human neurons. Sci Rep. 2015;5:10745.CrossRefPubMedPubMedCentral
37.
go back to reference Hirano M, Yoshii K, Sakai M, Hasebe R, Ichii O, Kariwa H. Tick-borne flaviviruses alter membrane structure and replicate in dendrites of primary mouse neuronal cultures. J Gen Virol. 2014;95:849–61.CrossRefPubMed Hirano M, Yoshii K, Sakai M, Hasebe R, Ichii O, Kariwa H. Tick-borne flaviviruses alter membrane structure and replicate in dendrites of primary mouse neuronal cultures. J Gen Virol. 2014;95:849–61.CrossRefPubMed
38.
go back to reference Růžek D, Vancová M, Tesařová M, Ahantarig A, Kopecký J, Grubhoffer L. Morphological changes in human neural cells following tick-borne encephalitis virus infection. J Gen Virol. 2009;90:1649–58.CrossRefPubMed Růžek D, Vancová M, Tesařová M, Ahantarig A, Kopecký J, Grubhoffer L. Morphological changes in human neural cells following tick-borne encephalitis virus infection. J Gen Virol. 2009;90:1649–58.CrossRefPubMed
39.
go back to reference Süss J, Gelpi E, Klaus C, Bagon A, Liebler-Tenorio EM, Budka H, et al. Tickborne encephalitis in naturally exposed monkey (Macaca sylvanus). Emerg Infect Dis. 2007;13(6):905–7.CrossRefPubMedPubMedCentral Süss J, Gelpi E, Klaus C, Bagon A, Liebler-Tenorio EM, Budka H, et al. Tickborne encephalitis in naturally exposed monkey (Macaca sylvanus). Emerg Infect Dis. 2007;13(6):905–7.CrossRefPubMedPubMedCentral
40.
go back to reference Dorrbecker B, Dobler G, Spiegel M, Hufert FT. Tick-borne encephalitis virus and the immune response of the mammalian host. Travel Med Infect Dis. 2010;8(4):213–22.CrossRefPubMed Dorrbecker B, Dobler G, Spiegel M, Hufert FT. Tick-borne encephalitis virus and the immune response of the mammalian host. Travel Med Infect Dis. 2010;8(4):213–22.CrossRefPubMed
41.
go back to reference Růžek D, Salát J, Singh SK, Kopecký J. Breakdown of the blood-brain barrier during tick-borne encephalitis in mice is not dependent on CD8+ T-cells. PLoS ONE. 2011;6(5):e20472.CrossRefPubMedPubMedCentral Růžek D, Salát J, Singh SK, Kopecký J. Breakdown of the blood-brain barrier during tick-borne encephalitis in mice is not dependent on CD8+ T-cells. PLoS ONE. 2011;6(5):e20472.CrossRefPubMedPubMedCentral
42.
go back to reference Tigabu B, Juelich T, Holbrook MR. Comparative analysis of immune responses to Russian spring–summer encephalitis and Omsk hemorrhagic fever viruses in mouse models. Virology. 2010;408(1):57–63.CrossRefPubMedPubMedCentral Tigabu B, Juelich T, Holbrook MR. Comparative analysis of immune responses to Russian spring–summer encephalitis and Omsk hemorrhagic fever viruses in mouse models. Virology. 2010;408(1):57–63.CrossRefPubMedPubMedCentral
43.
go back to reference Ridge JP, Fuchs EJ, Matzinger P. Neonatal tolerance revisited: turning on newborn T cells with dendritic cells. Science. 1996;271(5256):1723–6.CrossRefPubMed Ridge JP, Fuchs EJ, Matzinger P. Neonatal tolerance revisited: turning on newborn T cells with dendritic cells. Science. 1996;271(5256):1723–6.CrossRefPubMed
44.
go back to reference Lambert P-H, Liu M, Siegrist C-A. Can successful vaccines teach us how to induce efficient protective immune responses? Nat Med. 2005;11:S54–62.CrossRefPubMed Lambert P-H, Liu M, Siegrist C-A. Can successful vaccines teach us how to induce efficient protective immune responses? Nat Med. 2005;11:S54–62.CrossRefPubMed
45.
go back to reference Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol. 2004;4(7):553–64.CrossRefPubMed Adkins B, Leclerc C, Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol. 2004;4(7):553–64.CrossRefPubMed
46.
go back to reference Feuer R, Ruller CM, An N, Tabor-Godwin JM, Rhoades RE, Maciejewski S, et al. Viral persistence and chronic immunopathology in the adult central nervous system following Coxsackievirus infection during the neonatal period. J Virol. 2009;83(18):9356–69.CrossRefPubMedPubMedCentral Feuer R, Ruller CM, An N, Tabor-Godwin JM, Rhoades RE, Maciejewski S, et al. Viral persistence and chronic immunopathology in the adult central nervous system following Coxsackievirus infection during the neonatal period. J Virol. 2009;83(18):9356–69.CrossRefPubMedPubMedCentral
47.
go back to reference Lines JL, Hoskins S, Hollifield M, Cauley LS, Garvy BA. The migration of T cells in response to influenza virus is altered in neonatal mice. J Immunol. 2010;185(5):2980–8.CrossRefPubMedPubMedCentral Lines JL, Hoskins S, Hollifield M, Cauley LS, Garvy BA. The migration of T cells in response to influenza virus is altered in neonatal mice. J Immunol. 2010;185(5):2980–8.CrossRefPubMedPubMedCentral
48.
go back to reference Lacroix-Lamande S, Mancassola R, Naciri M, Laurent F. Role of gamma interferon in chemokine expression in the ileum of mice and in a murine intestinal epithelial cell line after Cryptosporidium parvum infection. Infect Immun. 2002;70(4):2090–9.CrossRefPubMedPubMedCentral Lacroix-Lamande S, Mancassola R, Naciri M, Laurent F. Role of gamma interferon in chemokine expression in the ileum of mice and in a murine intestinal epithelial cell line after Cryptosporidium parvum infection. Infect Immun. 2002;70(4):2090–9.CrossRefPubMedPubMedCentral
49.
go back to reference Wallner G, Mandl CW, Ecker M, Holzmann H, Stiasny K, Kunz C, et al. Characterization and complete genome sequences of high- and low-virulence variants of tick-borne encephalitis virus. J Gen Virol. 1996;77(Pt 5):1035–42.CrossRefPubMed Wallner G, Mandl CW, Ecker M, Holzmann H, Stiasny K, Kunz C, et al. Characterization and complete genome sequences of high- and low-virulence variants of tick-borne encephalitis virus. J Gen Virol. 1996;77(Pt 5):1035–42.CrossRefPubMed
50.
go back to reference Chiba N, Iwasaki T, Mizutani T, Kariwa H, Kurata T, Takashima I. Pathogenicity of tick-borne encephalitis virus isolated in Hokkaido, Japan in mouse model. Vaccine. 1999;17(7-8):779–87.CrossRefPubMed Chiba N, Iwasaki T, Mizutani T, Kariwa H, Kurata T, Takashima I. Pathogenicity of tick-borne encephalitis virus isolated in Hokkaido, Japan in mouse model. Vaccine. 1999;17(7-8):779–87.CrossRefPubMed
51.
go back to reference Appay V, Rowland-Jones SL. RANTES: a versatile and controversial chemokine. Trends Immunol. 2001;22(2):83–7.CrossRefPubMed Appay V, Rowland-Jones SL. RANTES: a versatile and controversial chemokine. Trends Immunol. 2001;22(2):83–7.CrossRefPubMed
52.
go back to reference Gangwani MR, Noel Jr RJ, Shah A, Rivera-Amill V, Kumar A. Human immunodeficiency virus type 1 viral protein R (Vpr) induces CCL5 expression in astrocytes via PI-3 K and MAPK signaling pathways. J Neuroinflammation. 2013;10:136.CrossRefPubMedPubMedCentral Gangwani MR, Noel Jr RJ, Shah A, Rivera-Amill V, Kumar A. Human immunodeficiency virus type 1 viral protein R (Vpr) induces CCL5 expression in astrocytes via PI-3 K and MAPK signaling pathways. J Neuroinflammation. 2013;10:136.CrossRefPubMedPubMedCentral
53.
go back to reference Chen C-J, Chen J-H, Chen S-Y, Liao S-L, Raung S-L. Upregulation of RANTES gene expression in neuroglia by Japanese encephalitis virus infection. J Virol. 2004;78(22):12107–19.CrossRefPubMedPubMedCentral Chen C-J, Chen J-H, Chen S-Y, Liao S-L, Raung S-L. Upregulation of RANTES gene expression in neuroglia by Japanese encephalitis virus infection. J Virol. 2004;78(22):12107–19.CrossRefPubMedPubMedCentral
54.
go back to reference Vilela MC, Mansur DS, Lacerda‐Queiroz N, Rodrigues DH, Lima GK, Arantes RME, et al. The chemokine CCL5 is essential for leukocyte recruitment in a model of severe herpes simplex encephalitis. Ann NY Acad Sci. 2009;1153(1):256–63.CrossRefPubMed Vilela MC, Mansur DS, Lacerda‐Queiroz N, Rodrigues DH, Lima GK, Arantes RME, et al. The chemokine CCL5 is essential for leukocyte recruitment in a model of severe herpes simplex encephalitis. Ann NY Acad Sci. 2009;1153(1):256–63.CrossRefPubMed
55.
go back to reference Lane TE, Liu MT, Chen BP, Asensio VC, Samawi RM, Paoletti AD, et al. A central role for CD4+ T cells and RANTES in virus-induced central nervous system inflammation and demyelination. J Virol. 2000;74(3):1415–24.CrossRefPubMedPubMedCentral Lane TE, Liu MT, Chen BP, Asensio VC, Samawi RM, Paoletti AD, et al. A central role for CD4+ T cells and RANTES in virus-induced central nervous system inflammation and demyelination. J Virol. 2000;74(3):1415–24.CrossRefPubMedPubMedCentral
56.
go back to reference Huang Y, Jiao S, Tao X, Tang Q, Jiao W, Xiao J, et al. Met-CCL5 represents an immunotherapy strategy to ameliorate rabies virus infection. J Neuroinflammation. 2014;11:146.CrossRefPubMedPubMedCentral Huang Y, Jiao S, Tao X, Tang Q, Jiao W, Xiao J, et al. Met-CCL5 represents an immunotherapy strategy to ameliorate rabies virus infection. J Neuroinflammation. 2014;11:146.CrossRefPubMedPubMedCentral
57.
go back to reference Glass WG, Lim JK, Cholera R, Pletnev AG, Gao J-L, Murphy PM. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection. J Exp Med. 2005;202(8):1087–98.CrossRefPubMedPubMedCentral Glass WG, Lim JK, Cholera R, Pletnev AG, Gao J-L, Murphy PM. Chemokine receptor CCR5 promotes leukocyte trafficking to the brain and survival in West Nile virus infection. J Exp Med. 2005;202(8):1087–98.CrossRefPubMedPubMedCentral
58.
go back to reference Larena M, Regner M, Lobigs M. The chemokine receptor CCR5, a therapeutic target for HIV/AIDS antagonists, is critical for recovery in a mouse model of Japanese encephalitis. PLoS ONE. 2012;7:e44834.CrossRefPubMedPubMedCentral Larena M, Regner M, Lobigs M. The chemokine receptor CCR5, a therapeutic target for HIV/AIDS antagonists, is critical for recovery in a mouse model of Japanese encephalitis. PLoS ONE. 2012;7:e44834.CrossRefPubMedPubMedCentral
59.
go back to reference Kindberg E, Mickienė A, Ax C, Åkerlind B, Vene S, Lindquist L, et al. A deletion in the chemokine receptor 5 (CCR5) gene is associated with tickborne encephalitis. J Infect Dis. 2008;197(2):266–9.CrossRefPubMed Kindberg E, Mickienė A, Ax C, Åkerlind B, Vene S, Lindquist L, et al. A deletion in the chemokine receptor 5 (CCR5) gene is associated with tickborne encephalitis. J Infect Dis. 2008;197(2):266–9.CrossRefPubMed
60.
go back to reference Mickiene A, Pakalniene J, Nordgren J, Carlsson B, Hagbom M, Svensson L, et al. Polymorphisms in chemokine receptor 5 and Toll-like receptor 3 genes are risk factors for clinical tick-borne encephalitis in the Lithuanian population. PLoS ONE. 2014;9(9):e106798.CrossRefPubMedPubMedCentral Mickiene A, Pakalniene J, Nordgren J, Carlsson B, Hagbom M, Svensson L, et al. Polymorphisms in chemokine receptor 5 and Toll-like receptor 3 genes are risk factors for clinical tick-borne encephalitis in the Lithuanian population. PLoS ONE. 2014;9(9):e106798.CrossRefPubMedPubMedCentral
61.
go back to reference Barkhash AV, Voevoda MI, Romaschenko AG. Association of single nucleotide polymorphism rs3775291 in the coding region of the TLR3 gene with predisposition to tick-borne encephalitis in a Russian population. Antiviral Res. 2013;99(2):136–8.CrossRefPubMed Barkhash AV, Voevoda MI, Romaschenko AG. Association of single nucleotide polymorphism rs3775291 in the coding region of the TLR3 gene with predisposition to tick-borne encephalitis in a Russian population. Antiviral Res. 2013;99(2):136–8.CrossRefPubMed
62.
go back to reference Overby AK, Popov VL, Niedrig M, Weber F. Tick-borne encephalitis virus delays interferon induction and hides its double-stranded RNA in intracellular membrane vesicles. J Virol. 2010;84(17):8470–83.CrossRefPubMedPubMedCentral Overby AK, Popov VL, Niedrig M, Weber F. Tick-borne encephalitis virus delays interferon induction and hides its double-stranded RNA in intracellular membrane vesicles. J Virol. 2010;84(17):8470–83.CrossRefPubMedPubMedCentral
63.
go back to reference Miorin L, Albornoz A, Baba MM, D’Agaro P, Marcello A. Formation of membrane-defined compartments by tick-borne encephalitis virus contributes to the early delay in interferon signaling. Virus Res. 2012;163(2):660–6.CrossRefPubMed Miorin L, Albornoz A, Baba MM, D’Agaro P, Marcello A. Formation of membrane-defined compartments by tick-borne encephalitis virus contributes to the early delay in interferon signaling. Virus Res. 2012;163(2):660–6.CrossRefPubMed
64.
go back to reference Melchjorsen J, Sørensen LN, Paludan SR. Expression and function of chemokines during viral infections: from molecular mechanisms to in vivo function. J Leukocyte Biol. 2003;74(3):331–43.CrossRefPubMed Melchjorsen J, Sørensen LN, Paludan SR. Expression and function of chemokines during viral infections: from molecular mechanisms to in vivo function. J Leukocyte Biol. 2003;74(3):331–43.CrossRefPubMed
65.
go back to reference Song A, Nikolcheva T, Krensky AM. Transcriptional regulation of RANTES expression in T lymphocytes. Immunol Rev. 2000;177(1):236–45.CrossRefPubMed Song A, Nikolcheva T, Krensky AM. Transcriptional regulation of RANTES expression in T lymphocytes. Immunol Rev. 2000;177(1):236–45.CrossRefPubMed
66.
go back to reference Lin R, Heylbroeck C, Genin P, Pitha PM, Hiscott J. Essential role of interferon regulatory factor 3 in direct activation of RANTES chemokine transcription. Mol Cell Biol. 1999;19(2):959–66.CrossRefPubMedPubMedCentral Lin R, Heylbroeck C, Genin P, Pitha PM, Hiscott J. Essential role of interferon regulatory factor 3 in direct activation of RANTES chemokine transcription. Mol Cell Biol. 1999;19(2):959–66.CrossRefPubMedPubMedCentral
67.
go back to reference Melchjorsen J, Paludan SR. Induction of RANTES/CCL5 by herpes simplex virus is regulated by nuclear factor kB and interferon regulatory factor 3. J Gen Virol. 2003;84(9):2491–5.CrossRefPubMed Melchjorsen J, Paludan SR. Induction of RANTES/CCL5 by herpes simplex virus is regulated by nuclear factor kB and interferon regulatory factor 3. J Gen Virol. 2003;84(9):2491–5.CrossRefPubMed
68.
go back to reference Wang Y, Luo R, Fang L, Wang D, Bi J, Chen H, et al. Porcine reproductive and respiratory syndrome virus (PRRSV) infection activates chemokine RANTES in MARC-145 cells. Mol Immunol. 2011;48(4):586–91.CrossRefPubMed Wang Y, Luo R, Fang L, Wang D, Bi J, Chen H, et al. Porcine reproductive and respiratory syndrome virus (PRRSV) infection activates chemokine RANTES in MARC-145 cells. Mol Immunol. 2011;48(4):586–91.CrossRefPubMed
69.
go back to reference Lin Y-L, Liu C-C, Chuang J-I, Lei H-Y, Yeh T-M, Lin Y-S, et al. Involvement of oxidative stress, NF-IL-6, and RANTES expression in dengue-2-virus-infected human liver cells. Virology. 2000;276(1):114–26.CrossRefPubMed Lin Y-L, Liu C-C, Chuang J-I, Lei H-Y, Yeh T-M, Lin Y-S, et al. Involvement of oxidative stress, NF-IL-6, and RANTES expression in dengue-2-virus-infected human liver cells. Virology. 2000;276(1):114–26.CrossRefPubMed
70.
go back to reference Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441(7089):101–5.CrossRefPubMed Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441(7089):101–5.CrossRefPubMed
71.
go back to reference Errett JS, Suthar MS, McMillan A, Diamond MS, Gale Jr M. The essential, nonredundant roles of RIG-I and MDA5 in detecting and controlling West Nile virus infection. J Virol. 2013;87(21):11416–25.CrossRefPubMedPubMedCentral Errett JS, Suthar MS, McMillan A, Diamond MS, Gale Jr M. The essential, nonredundant roles of RIG-I and MDA5 in detecting and controlling West Nile virus infection. J Virol. 2013;87(21):11416–25.CrossRefPubMedPubMedCentral
72.
go back to reference Mesman AW, Zijlstra-Willems EM, Kaptein TM, de Swart RL, Davis ME, Ludlow M, et al. Measles virus suppresses RIG-I-like receptor activation in dendritic cells via DC-SIGN-mediated inhibition of PP1 phosphatases. Cell Host Microbe. 2014;16(1):31–42.CrossRefPubMedPubMedCentral Mesman AW, Zijlstra-Willems EM, Kaptein TM, de Swart RL, Davis ME, Ludlow M, et al. Measles virus suppresses RIG-I-like receptor activation in dendritic cells via DC-SIGN-mediated inhibition of PP1 phosphatases. Cell Host Microbe. 2014;16(1):31–42.CrossRefPubMedPubMedCentral
73.
go back to reference Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E, Golenbock DT, et al. IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol. 2003;4(5):491–6.CrossRefPubMed Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E, Golenbock DT, et al. IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol. 2003;4(5):491–6.CrossRefPubMed
74.
go back to reference Sharma S, tenOever BR, Grandvaux N, Zhou GP, Lin R, Hiscott J. Triggering the interferon antiviral response through an IKK-related pathway. Science. 2003;300(5622):1148–51.CrossRefPubMed Sharma S, tenOever BR, Grandvaux N, Zhou GP, Lin R, Hiscott J. Triggering the interferon antiviral response through an IKK-related pathway. Science. 2003;300(5622):1148–51.CrossRefPubMed
75.
go back to reference Genin P, Algarte M, Roof P, Lin R, Hiscott J. Regulation of RANTES chemokine gene expression requires cooperativity between NF-kappa B and IFN-regulatory factor transcription factors. J Immunol. 2000;164(10):5352–61.CrossRefPubMed Genin P, Algarte M, Roof P, Lin R, Hiscott J. Regulation of RANTES chemokine gene expression requires cooperativity between NF-kappa B and IFN-regulatory factor transcription factors. J Immunol. 2000;164(10):5352–61.CrossRefPubMed
Metadata
Title
Tick-borne encephalitis virus induces chemokine RANTES expression via activation of IRF-3 pathway
Authors
Xiaowei Zhang
Zhenhua Zheng
Xijuan Liu
Bo Shu
Panyong Mao
Bingke Bai
Qinxue Hu
Minhua Luo
Xiaohe Ma
Zongqiang Cui
Hanzhong Wang
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0665-9

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue