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

Open Access 01-12-2016 | Research

CCR5 ameliorates Japanese encephalitis via dictating the equilibrium of regulatory CD4+Foxp3+ T and IL-17+CD4+ Th17 cells

Authors: Jin Hyoung Kim, Ajit Mahadev Patil, Jin Young Choi, Seong Bum Kim, Erdenebelig Uyangaa, Ferdaus Mohd Altaf Hossain, Sang-Youel Park, John Hwa Lee, Seong Kug Eo

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

CCR5 is a CC chemokine receptor involved in the migration of effector leukocytes including macrophages, NK, and T cells into inflamed tissues. Also, the role of CCR5 in CD4+Foxp3+ regulatory T cell (Treg) homing has recently begun to grab attention. Japanese encephalitis (JE) is defined as severe neuroinflammation of the central nervous system (CNS) following infection with mosquito-borne flavivirus JE virus. However, the potential contribution of CCR5 to JE progression via mediating CD4+Foxp3+ Treg homing has not been investigated.

Methods

Infected wild-type (Ccr5+/+) and CCR5-deficient (Ccr5−/−) mice were examined daily for mortality and clinical signs, and neuroinflammation in the CNS was evaluated by infiltration of inflammatory leukocytes and cytokine expression. In addition, viral burden, NK- and JEV-specific T cell responses were analyzed. Adoptive transfer of CCR5+CD4+Foxp3+ Tregs was used to evaluate the role of Tregs in JE progression.

Results

CCR5 ablation exacerbated JE without altering viral burden in the extraneural and CNS tissues, as manifested by increased CNS infiltration of Ly-6Chi monocytes and Ly-6Ghi granulocytes. Compared to Ccr5+/+ mice, Ccr5−/− mice unexpectedly showed increased responses of IFN-γ+NK and CD8+ T cells in the spleen, but not CD4+ T cells. More interestingly, CCR5-ablation resulted in a skewed response to IL-17+CD4+ Th17 cells and correspondingly reduced CD4+Foxp3+ Tregs in the spleen and brain, which was closely associated with exacerbated JE. Our results also revealed that adoptive transfer of sorted CCR5+CD4+Foxp3+ Tregs into Ccr5−/− mice could ameliorate JE progression without apparently altering the viral burden and CNS infiltration of IL-17+CD4+ Th17 cells, myeloid-derived Ly-6Chi monocytes and Ly-6Ghi granulocytes. Instead, adoptive transfer of CCR5+CD4+Foxp3+ Tregs into Ccr5−/− mice resulted in increased expression of anti-inflammatory cytokines (IL-10 and TGF-β) in the spleen and brain, and transferred CCR5+ Tregs were found to produce IL-10.

Conclusions

CCR5 regulates JE progression via governing timely and appropriate CNS infiltration of CD4+Foxp3+ Tregs, thereby facilitating host survival. Therefore, this critical and extended role of CCR5 in JE raises possible safety concerns regarding the use of CCR5 antagonists in human immunodeficiency virus (HIV)-infected individuals who inhabit regions in which both HIV and flaviviruses, such as JEV and West Nile virus, are endemic.
Literature
1.
go back to reference Sips GJ, Wilschut J, Smit JM. Neuroinvasive flavivirus infections. Rev Med Virol. 2012;22(2):69–87.PubMedCrossRef Sips GJ, Wilschut J, Smit JM. Neuroinvasive flavivirus infections. Rev Med Virol. 2012;22(2):69–87.PubMedCrossRef
3.
go back to reference Go YY, Balasuriya UB, Lee CK. Zoonotic encephalitides caused by arboviruses: transmission and epidemiology of alphaviruses and flaviviruses. Clin Exp Vaccine Res. 2014;3(1):58–77.PubMedCrossRef Go YY, Balasuriya UB, Lee CK. Zoonotic encephalitides caused by arboviruses: transmission and epidemiology of alphaviruses and flaviviruses. Clin Exp Vaccine Res. 2014;3(1):58–77.PubMedCrossRef
4.
go back to reference Japanese encephalitis: status of surveillance and immunization in Asia and the Western Pacific, 2012. Wkly Epidemiol Rec. 2013;88(34):357-364. Japanese encephalitis: status of surveillance and immunization in Asia and the Western Pacific, 2012. Wkly Epidemiol Rec. 2013;88(34):357-364.
5.
go back to reference Mackenzie JS, Gubler DJ, Petersen LR. Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat Med. 2004;10(12 Suppl):S98–109.PubMedCrossRef Mackenzie JS, Gubler DJ, Petersen LR. Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nat Med. 2004;10(12 Suppl):S98–109.PubMedCrossRef
7.
go back to reference King NJ, Getts DR, Getts MT, Rana S, Shrestha B, Kesson AM. Immunopathology of flavivirus infections. Immunol Cell Biol. 2007;85(1):33–42.PubMedCrossRef King NJ, Getts DR, Getts MT, Rana S, Shrestha B, Kesson AM. Immunopathology of flavivirus infections. Immunol Cell Biol. 2007;85(1):33–42.PubMedCrossRef
8.
go back to reference Kimura T, Sasaki M, Okumura M, Kim E, Sawa H. Flavivirus encephalitis: pathological aspects of mouse and other animal models. Vet Pathol. 2010;47(5):806–18.PubMedCrossRef Kimura T, Sasaki M, Okumura M, Kim E, Sawa H. Flavivirus encephalitis: pathological aspects of mouse and other animal models. Vet Pathol. 2010;47(5):806–18.PubMedCrossRef
9.
go back to reference Han YW, Singh SK, Eo SK. The roles and perspectives of toll-like receptors and CD4(+) helper T cell subsets in acute viral encephalitis. Immune Netw. 2012;12(2):48–57.PubMedPubMedCentralCrossRef Han YW, Singh SK, Eo SK. The roles and perspectives of toll-like receptors and CD4(+) helper T cell subsets in acute viral encephalitis. Immune Netw. 2012;12(2):48–57.PubMedPubMedCentralCrossRef
10.
go back to reference Chen CJ, Ou YC, Lin SY, Raung SL, Liao SL, Lai CY, et al. Glial activation involvement in neuronal death by Japanese encephalitis virus infection. J Gen Virol. 2010;91(Pt 4):1028–37.PubMedCrossRef Chen CJ, Ou YC, Lin SY, Raung SL, Liao SL, Lai CY, et al. Glial activation involvement in neuronal death by Japanese encephalitis virus infection. J Gen Virol. 2010;91(Pt 4):1028–37.PubMedCrossRef
11.
go back to reference Ghoshal A, Das S, Ghosh S, Mishra MK, Sharma V, Koli P, et al. Proinflammatory mediators released by activated microglia induces neuronal death in Japanese encephalitis. Glia. 2007;55(5):483–96.PubMedCrossRef Ghoshal A, Das S, Ghosh S, Mishra MK, Sharma V, Koli P, et al. Proinflammatory mediators released by activated microglia induces neuronal death in Japanese encephalitis. Glia. 2007;55(5):483–96.PubMedCrossRef
12.
go back to reference Bardina SV, Lim JK. The role of chemokines in the pathogenesis of neurotropic flaviviruses. Immunol Res. 2012;54(1-3):121–32.PubMedCrossRef Bardina SV, Lim JK. The role of chemokines in the pathogenesis of neurotropic flaviviruses. Immunol Res. 2012;54(1-3):121–32.PubMedCrossRef
13.
go back to reference Zhang B, Chan YK, Lu B, Diamond MS, Klein RS. CXCR3 mediates region-specific antiviral T cell trafficking within the central nervous system during West Nile virus encephalitis. J Immunol. 2008;180(4):2641–9.PubMedCrossRef Zhang B, Chan YK, Lu B, Diamond MS, Klein RS. CXCR3 mediates region-specific antiviral T cell trafficking within the central nervous system during West Nile virus encephalitis. J Immunol. 2008;180(4):2641–9.PubMedCrossRef
14.
go back to reference Hofer MJ, Carter SL, Muller M, Campbell IL. Unaltered neurological disease and mortality in CXCR3-deficient mice infected intracranially with lymphocytic choriomeningitis virus-Armstrong. Viral Immunol. 2008;21(4):425–33.PubMedPubMedCentralCrossRef Hofer MJ, Carter SL, Muller M, Campbell IL. Unaltered neurological disease and mortality in CXCR3-deficient mice infected intracranially with lymphocytic choriomeningitis virus-Armstrong. Viral Immunol. 2008;21(4):425–33.PubMedPubMedCentralCrossRef
15.
go back to reference Campanella GS, Tager AM, El Khoury JK, Thomas SY, Abrazinski TA, Manice LA, et al. Chemokine receptor CXCR3 and its ligands CXCL9 and CXCL10 are required for the development of murine cerebral malaria. Proc Natl Acd Sci USA. 2008;105(12):4814–9.CrossRef Campanella GS, Tager AM, El Khoury JK, Thomas SY, Abrazinski TA, Manice LA, et al. Chemokine receptor CXCR3 and its ligands CXCL9 and CXCL10 are required for the development of murine cerebral malaria. Proc Natl Acd Sci USA. 2008;105(12):4814–9.CrossRef
16.
go back to reference Glass WG, Lim JK, Cholera R, Pletnev AG, Gao JL, 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.PubMedPubMedCentralCrossRef Glass WG, Lim JK, Cholera R, Pletnev AG, Gao JL, 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.PubMedPubMedCentralCrossRef
17.
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(9):e44834.PubMedPubMedCentralCrossRef 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(9):e44834.PubMedPubMedCentralCrossRef
18.
go back to reference Michlmayr D, McKimmie CS, Pingen M, Haxton B, Mansfield K, Johnson N, et al. Defining the chemokine basis for leukocyte recruitment during viral encephalitis. J Virol. 2014;88(17):9553–67.PubMedPubMedCentralCrossRef Michlmayr D, McKimmie CS, Pingen M, Haxton B, Mansfield K, Johnson N, et al. Defining the chemokine basis for leukocyte recruitment during viral encephalitis. J Virol. 2014;88(17):9553–67.PubMedPubMedCentralCrossRef
19.
go back to reference Glass WG, McDermott DH, Lim JK, Lekhong S, Yu SF, Frank WA, et al. CCR5 deficiency increases risk of symptomatic West Nile virus infection. J Exp Med. 2006;203(1):35–40.PubMedPubMedCentralCrossRef Glass WG, McDermott DH, Lim JK, Lekhong S, Yu SF, Frank WA, et al. CCR5 deficiency increases risk of symptomatic West Nile virus infection. J Exp Med. 2006;203(1):35–40.PubMedPubMedCentralCrossRef
20.
go back to reference Zimmerman PA, Buckler-White A, Alkhatib G, Spalding T, Kubofcik J, Combadiere C, et al. Inherited resistance to HIV-1 conferred by an inactivating mutation in CC chemokine receptor 5: studies in populations with contrasting clinical phenotypes, defined racial background, and quantified risk. Mol Med. 1997;3(1):23–36.PubMedPubMedCentral Zimmerman PA, Buckler-White A, Alkhatib G, Spalding T, Kubofcik J, Combadiere C, et al. Inherited resistance to HIV-1 conferred by an inactivating mutation in CC chemokine receptor 5: studies in populations with contrasting clinical phenotypes, defined racial background, and quantified risk. Mol Med. 1997;3(1):23–36.PubMedPubMedCentral
23.
go back to reference Moreira AP, Cavassani KA, Massafera Tristao FS, Campanelli AP, Martinez R, Rossi MA, et al. CCR5-dependent regulatory T cell migration mediates fungal survival and severe immunosuppression. J Immunol. 2008;180(5):3049–56.PubMedCrossRef Moreira AP, Cavassani KA, Massafera Tristao FS, Campanelli AP, Martinez R, Rossi MA, et al. CCR5-dependent regulatory T cell migration mediates fungal survival and severe immunosuppression. J Immunol. 2008;180(5):3049–56.PubMedCrossRef
24.
go back to reference Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. J Exp Med. 2006;203(11):2451–60.PubMedPubMedCentralCrossRef Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. J Exp Med. 2006;203(11):2451–60.PubMedPubMedCentralCrossRef
25.
go back to reference Lanteri MC, O'Brien KM, Purtha WE, Cameron MJ, Lund JM, Owen RE, et al. Tregs control the development of symptomatic West Nile virus infection in humans and mice. J Clin Invest. 2009;119(11):3266–77.PubMedPubMedCentral Lanteri MC, O'Brien KM, Purtha WE, Cameron MJ, Lund JM, Owen RE, et al. Tregs control the development of symptomatic West Nile virus infection in humans and mice. J Clin Invest. 2009;119(11):3266–77.PubMedPubMedCentral
26.
go back to reference Luhn K, Simmons CP, Moran E, Dung NT, Chau TN, Quyen NT, et al. Increased frequencies of CD4+ CD25(high) regulatory T cells in acute dengue infection. J Exp Med. 2007;204(5):979–85.PubMedPubMedCentralCrossRef Luhn K, Simmons CP, Moran E, Dung NT, Chau TN, Quyen NT, et al. Increased frequencies of CD4+ CD25(high) regulatory T cells in acute dengue infection. J Exp Med. 2007;204(5):979–85.PubMedPubMedCentralCrossRef
27.
go back to reference Han YW, Choi JY, Uyangaa E, Kim SB, Kim JH, Kim BS, et al. Distinct dictation of Japanese encephalitis virus-induced neuroinflammation and lethality via triggering TLR3 and TLR4 signal pathways. PLoS Pathog. 2014;10(9):e1004319.PubMedPubMedCentralCrossRef Han YW, Choi JY, Uyangaa E, Kim SB, Kim JH, Kim BS, et al. Distinct dictation of Japanese encephalitis virus-induced neuroinflammation and lethality via triggering TLR3 and TLR4 signal pathways. PLoS Pathog. 2014;10(9):e1004319.PubMedPubMedCentralCrossRef
28.
go back to reference Frentsch M, Arbach O, Kirchhoff D, Moewes B, Worm M, Rothe M, et al. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat Med. 2005;11(10):1118–24.PubMedCrossRef Frentsch M, Arbach O, Kirchhoff D, Moewes B, Worm M, Rothe M, et al. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat Med. 2005;11(10):1118–24.PubMedCrossRef
29.
go back to reference Chattopadhyay PK, Yu J, Roederer M. Live-cell assay to detect antigen-specific CD4+ T-cell responses by CD154 expression. Nat Protoc. 2006;1(1):1–6.PubMedCrossRef Chattopadhyay PK, Yu J, Roederer M. Live-cell assay to detect antigen-specific CD4+ T-cell responses by CD154 expression. Nat Protoc. 2006;1(1):1–6.PubMedCrossRef
30.
go back to reference Terry RL, Getts DR, Deffrasnes C, van Vreden C, Campbell IL, King NJ. Inflammatory monocytes and the pathogenesis of viral encephalitis. J Neuroinflammation. 2012;9:270.PubMedPubMedCentralCrossRef Terry RL, Getts DR, Deffrasnes C, van Vreden C, Campbell IL, King NJ. Inflammatory monocytes and the pathogenesis of viral encephalitis. J Neuroinflammation. 2012;9:270.PubMedPubMedCentralCrossRef
31.
go back to reference Getts DR, Terry RL, Getts MT, Muller M, Rana S, Shrestha B, et al. Ly6c+ “inflammatory monocytes” are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J Exp Med. 2008;205(10):2319–37.PubMedPubMedCentralCrossRef Getts DR, Terry RL, Getts MT, Muller M, Rana S, Shrestha B, et al. Ly6c+ “inflammatory monocytes” are microglial precursors recruited in a pathogenic manner in West Nile virus encephalitis. J Exp Med. 2008;205(10):2319–37.PubMedPubMedCentralCrossRef
32.
go back to reference Suzumura A. Neuron-microglia interaction in neuroinflammation. Curr Protein Pept Sci. 2013;14(1):16–20.PubMedCrossRef Suzumura A. Neuron-microglia interaction in neuroinflammation. Curr Protein Pept Sci. 2013;14(1):16–20.PubMedCrossRef
33.
go back to reference Ford AL, Foulcher E, Lemckert FA, Sedgwick JD. Microglia induce CD4 T lymphocyte final effector function and death. J Exp Med. 1996;184(5):1737–45.PubMedCrossRef Ford AL, Foulcher E, Lemckert FA, Sedgwick JD. Microglia induce CD4 T lymphocyte final effector function and death. J Exp Med. 1996;184(5):1737–45.PubMedCrossRef
34.
go back to reference Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr Top Microbiol Immunol. 2010;341:37–58.PubMed Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr Top Microbiol Immunol. 2010;341:37–58.PubMed
35.
go back to reference de Oliveira CE, Oda JM, Losi Guembarovski R, de Oliveira KB, Ariza CB, Neto JS, et al. CC chemokine receptor 5: the interface of host immunity and cancer. Dis Markers. 2014;2014:126954.PubMedPubMedCentralCrossRef de Oliveira CE, Oda JM, Losi Guembarovski R, de Oliveira KB, Ariza CB, Neto JS, et al. CC chemokine receptor 5: the interface of host immunity and cancer. Dis Markers. 2014;2014:126954.PubMedPubMedCentralCrossRef
36.
go back to reference Larena M, Regner M, Lobigs M. Cytolytic effector pathways and IFN-gamma help protect against Japanese encephalitis. Eur J Immunol. 2013;43(7):1789–98.PubMedCrossRef Larena M, Regner M, Lobigs M. Cytolytic effector pathways and IFN-gamma help protect against Japanese encephalitis. Eur J Immunol. 2013;43(7):1789–98.PubMedCrossRef
37.
go back to reference Larena M, Regner M, Lee E, Lobigs M. Pivotal role of antibody and subsidiary contribution of CD8+ T cells to recovery from infection in a murine model of Japanese encephalitis. J Virol. 2011;85(11):5446–55.PubMedPubMedCentralCrossRef Larena M, Regner M, Lee E, Lobigs M. Pivotal role of antibody and subsidiary contribution of CD8+ T cells to recovery from infection in a murine model of Japanese encephalitis. J Virol. 2011;85(11):5446–55.PubMedPubMedCentralCrossRef
38.
go back to reference Kumar P, Sulochana P, Nirmala G, Chandrashekar R, Haridattatreya M, Satchidanandam V. Impaired T helper 1 function of nonstructural protein 3-specific T cells in Japanese patients with encephalitis with neurological sequelae. J Infect Dis. 2004;189(5):880–91.PubMedCrossRef Kumar P, Sulochana P, Nirmala G, Chandrashekar R, Haridattatreya M, Satchidanandam V. Impaired T helper 1 function of nonstructural protein 3-specific T cells in Japanese patients with encephalitis with neurological sequelae. J Infect Dis. 2004;189(5):880–91.PubMedCrossRef
39.
go back to reference Kroetz DN, Deepe Jr GS. CCR5 dictates the equilibrium of proinflammatory IL-17+ and regulatory Foxp3+ T cells in fungal infection. J Immunol. 2010;184(9):5224–31.PubMedPubMedCentralCrossRef Kroetz DN, Deepe Jr GS. CCR5 dictates the equilibrium of proinflammatory IL-17+ and regulatory Foxp3+ T cells in fungal infection. J Immunol. 2010;184(9):5224–31.PubMedPubMedCentralCrossRef
40.
go back to reference Moser EK, Hufford MM, Braciale TJ. Late engagement of CD86 after influenza virus clearance promotes recovery in a FoxP3+ regulatory T cell dependent manner. PLoS Pathog. 2014;10(8):e1004315.PubMedPubMedCentralCrossRef Moser EK, Hufford MM, Braciale TJ. Late engagement of CD86 after influenza virus clearance promotes recovery in a FoxP3+ regulatory T cell dependent manner. PLoS Pathog. 2014;10(8):e1004315.PubMedPubMedCentralCrossRef
41.
go back to reference Haque A, Best SE, Amante FH, Mustafah S, Desbarrieres L, de Labastida F, et al. CD4+ natural regulatory T cells prevent experimental cerebral malaria via CTLA-4 when expanded in vivo. PLoS Pathog. 2010;6(12):e1001221.PubMedPubMedCentralCrossRef Haque A, Best SE, Amante FH, Mustafah S, Desbarrieres L, de Labastida F, et al. CD4+ natural regulatory T cells prevent experimental cerebral malaria via CTLA-4 when expanded in vivo. PLoS Pathog. 2010;6(12):e1001221.PubMedPubMedCentralCrossRef
42.
go back to reference Hardison JL, Wrightsman RA, Carpenter PM, Kuziel WA, Lane TE, Manning JE. The CC chemokine receptor 5 is important in control of parasite replication and acute cardiac inflammation following infection with Trypanosoma cruzi. Infect Immun. 2006;74(1):135–43.PubMedPubMedCentralCrossRef Hardison JL, Wrightsman RA, Carpenter PM, Kuziel WA, Lane TE, Manning JE. The CC chemokine receptor 5 is important in control of parasite replication and acute cardiac inflammation following infection with Trypanosoma cruzi. Infect Immun. 2006;74(1):135–43.PubMedPubMedCentralCrossRef
43.
go back to reference Khan IA, Thomas SY, Moretto MM, Lee FS, Islam SA, Combe C, et al. CCR5 is essential for NK cell trafficking and host survival following Toxoplasma gondii infection. PLoS Pathog. 2006;2(6):e49.PubMedPubMedCentralCrossRef Khan IA, Thomas SY, Moretto MM, Lee FS, Islam SA, Combe C, et al. CCR5 is essential for NK cell trafficking and host survival following Toxoplasma gondii infection. PLoS Pathog. 2006;2(6):e49.PubMedPubMedCentralCrossRef
44.
go back to reference Thapa M, Kuziel WA, Carr DJ. Susceptibility of CCR5-deficient mice to genital herpes simplex virus type 2 is linked to NK cell mobilization. J Virol. 2007;81(8):3704–13.PubMedPubMedCentralCrossRef Thapa M, Kuziel WA, Carr DJ. Susceptibility of CCR5-deficient mice to genital herpes simplex virus type 2 is linked to NK cell mobilization. J Virol. 2007;81(8):3704–13.PubMedPubMedCentralCrossRef
45.
go back to reference Fadel SA, Bromley SK, Medoff BD, Luster AD. CXCR3-deficiency protects influenza-infected CCR5-deficient mice from mortality. Eur J Immunol. 2008;38(12):3376–87.PubMedPubMedCentralCrossRef Fadel SA, Bromley SK, Medoff BD, Luster AD. CXCR3-deficiency protects influenza-infected CCR5-deficient mice from mortality. Eur J Immunol. 2008;38(12):3376–87.PubMedPubMedCentralCrossRef
46.
go back to reference Algood HM, Flynn JL. CCR5-deficient mice control Mycobacterium tuberculosis infection despite increased pulmonary lymphocytic infiltration. J Immunol. 2004;173(5):3287–96.PubMedCrossRef Algood HM, Flynn JL. CCR5-deficient mice control Mycobacterium tuberculosis infection despite increased pulmonary lymphocytic infiltration. J Immunol. 2004;173(5):3287–96.PubMedCrossRef
47.
go back to reference Ank N, Petersen K, Malmgaard L, Mogensen SC, Paludan SR. Age-dependent role for CCR5 in antiviral host defense against herpes simplex virus type 2. J Virol. 2005;79(15):9831–41.PubMedPubMedCentralCrossRef Ank N, Petersen K, Malmgaard L, Mogensen SC, Paludan SR. Age-dependent role for CCR5 in antiviral host defense against herpes simplex virus type 2. J Virol. 2005;79(15):9831–41.PubMedPubMedCentralCrossRef
48.
go back to reference Zhao J, Perlman S. Virus-specific regulatory T cells ameliorate encephalitis by repressing effector T cell functions from priming to effector stages. PLoS Pathog. 2014;10(8):e1004279.PubMedPubMedCentralCrossRef Zhao J, Perlman S. Virus-specific regulatory T cells ameliorate encephalitis by repressing effector T cell functions from priming to effector stages. PLoS Pathog. 2014;10(8):e1004279.PubMedPubMedCentralCrossRef
49.
go back to reference Nansen A, Christensen JP, Andreasen SO, Bartholdy C, Christensen JE, Thomsen AR. The role of CC chemokine receptor 5 in antiviral immunity. Blood. 2002;99(4):1237–45.PubMedCrossRef Nansen A, Christensen JP, Andreasen SO, Bartholdy C, Christensen JE, Thomsen AR. The role of CC chemokine receptor 5 in antiviral immunity. Blood. 2002;99(4):1237–45.PubMedCrossRef
50.
go back to reference Peterson KE, Errett JS, Wei T, Dimcheff DE, Ransohoff R, Kuziel WA, et al. MCP-1 and CCR2 contribute to non-lymphocyte-mediated brain disease induced by Fr98 polytropic retrovirus infection in mice: role for astrocytes in retroviral neuropathogenesis. J Virol. 2004;78(12):6449–58.PubMedPubMedCentralCrossRef Peterson KE, Errett JS, Wei T, Dimcheff DE, Ransohoff R, Kuziel WA, et al. MCP-1 and CCR2 contribute to non-lymphocyte-mediated brain disease induced by Fr98 polytropic retrovirus infection in mice: role for astrocytes in retroviral neuropathogenesis. J Virol. 2004;78(12):6449–58.PubMedPubMedCentralCrossRef
51.
go back to reference Glass WG, Liu MT, Kuziel WA, Lane TE. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus. Virology. 2001;288(1):8–17.PubMedCrossRef Glass WG, Liu MT, Kuziel WA, Lane TE. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus. Virology. 2001;288(1):8–17.PubMedCrossRef
52.
go back to reference Kim JH, Choi JY, Kim SB, Uyangaa E, Patil AM, Han YW, et al. CD11chi dendritic cells regulate Ly-6Chi monocyte differentiation to preserve immune-privileged CNS in lethal neuroinflammation. Sci Rep. 2015;5:17548.PubMedPubMedCentralCrossRef Kim JH, Choi JY, Kim SB, Uyangaa E, Patil AM, Han YW, et al. CD11chi dendritic cells regulate Ly-6Chi monocyte differentiation to preserve immune-privileged CNS in lethal neuroinflammation. Sci Rep. 2015;5:17548.PubMedPubMedCentralCrossRef
53.
go back to reference Cazareth J, Guyon A, Heurteaux C, Chabry J, Petit-Paitel A. Molecular and cellular neuroinflammation status of mouse brain after systemic lipopolysaccharide challenge: importance of CCR2/CCL2 signaling. J Neuroinflammation. 2014;11:132.PubMedPubMedCentralCrossRef Cazareth J, Guyon A, Heurteaux C, Chabry J, Petit-Paitel A. Molecular and cellular neuroinflammation status of mouse brain after systemic lipopolysaccharide challenge: importance of CCR2/CCL2 signaling. J Neuroinflammation. 2014;11:132.PubMedPubMedCentralCrossRef
54.
go back to reference Reichel CA, Khandoga A, Anders HJ, Schlondorff D, Luckow B, Krombach F. Chemokine receptors Ccr1, Ccr2, and Ccr5 mediate neutrophul migration to postischemic tissue. J Leukoc Biol. 2006;79(1):114–22.PubMedCrossRef Reichel CA, Khandoga A, Anders HJ, Schlondorff D, Luckow B, Krombach F. Chemokine receptors Ccr1, Ccr2, and Ccr5 mediate neutrophul migration to postischemic tissue. J Leukoc Biol. 2006;79(1):114–22.PubMedCrossRef
55.
go back to reference Vilela MC, Lima GK, Rodrigues DH, Lacerda-Queiroz N, Pedroso VSP, Miranda AS, et al. Absence of CCR5 increases neutrophil recruitment in severe herpetic encephalitis. BMC Neurosci. 2013;14:19.PubMedPubMedCentralCrossRef Vilela MC, Lima GK, Rodrigues DH, Lacerda-Queiroz N, Pedroso VSP, Miranda AS, et al. Absence of CCR5 increases neutrophil recruitment in severe herpetic encephalitis. BMC Neurosci. 2013;14:19.PubMedPubMedCentralCrossRef
56.
go back to reference Kang X, Zhang X, Liu Z, Xu H, Wang T, He L, et al. CXCR2-mediated granulocytic myeloid-derived suppressor cells’ functional characterization and their role in maternal fetal interface. DNA Cell Biol. 2016;35(7):358–65.PubMedCrossRef Kang X, Zhang X, Liu Z, Xu H, Wang T, He L, et al. CXCR2-mediated granulocytic myeloid-derived suppressor cells’ functional characterization and their role in maternal fetal interface. DNA Cell Biol. 2016;35(7):358–65.PubMedCrossRef
57.
go back to reference Lim JK, Obara CJ, Rivollier A, Pletnev AG, Kelsall BL, Murphy PM. Chemokine receptor CCR2 is critical for monocyte accumulation and survival in West Nile virus encephalitis. J Immunol. 2011;186(1):471–8.PubMedCrossRef Lim JK, Obara CJ, Rivollier A, Pletnev AG, Kelsall BL, Murphy PM. Chemokine receptor CCR2 is critical for monocyte accumulation and survival in West Nile virus encephalitis. J Immunol. 2011;186(1):471–8.PubMedCrossRef
58.
go back to reference Cardona AE, Sasse ME, Liu L, Cardona SM, Mizutani M, Savarin C, et al. Scavenging roles of chemokine receptors: chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues. Blood. 2008;112:256–63.PubMedPubMedCentralCrossRef Cardona AE, Sasse ME, Liu L, Cardona SM, Mizutani M, Savarin C, et al. Scavenging roles of chemokine receptors: chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues. Blood. 2008;112:256–63.PubMedPubMedCentralCrossRef
59.
go back to reference Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659–702.PubMedCrossRef Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659–702.PubMedCrossRef
60.
go back to reference Singh RP, Hasan S, Sharma S, Nagra S, Yamaguchi DT, Wong DT, et al. Th17 cells in inflammation and autoimmunity. Autoimmun Rev. 2014;13(12):1174–81.PubMedCrossRef Singh RP, Hasan S, Sharma S, Nagra S, Yamaguchi DT, Wong DT, et al. Th17 cells in inflammation and autoimmunity. Autoimmun Rev. 2014;13(12):1174–81.PubMedCrossRef
61.
go back to reference Christiaansen AF, Knudson CJ, Weiss KA, Varga SM. The CD4 T cell response to respiratory syncytial virus infection. Immunol Res. 2014;59(1-3):109–17.PubMedCrossRef Christiaansen AF, Knudson CJ, Weiss KA, Varga SM. The CD4 T cell response to respiratory syncytial virus infection. Immunol Res. 2014;59(1-3):109–17.PubMedCrossRef
62.
go back to reference Wang Q, Zhou J, Zhang B, Tian Z, Tang J, Zheng Y, et al. Hepatitis B virus induces IL-23 production in antigen presenting cells and causes liver damage via the IL-23/IL-17 axis. PLoS Pathog. 2013;9(6):e1003410.PubMedPubMedCentralCrossRef Wang Q, Zhou J, Zhang B, Tian Z, Tang J, Zheng Y, et al. Hepatitis B virus induces IL-23 production in antigen presenting cells and causes liver damage via the IL-23/IL-17 axis. PLoS Pathog. 2013;9(6):e1003410.PubMedPubMedCentralCrossRef
63.
go back to reference Peckham RK, Brill R, Foster DS, Bowen AL, Leigh JA, Coffey TJ, et al. Two distinct populations of bovine IL-17+ T-cells can be induced and WC1+IL-17+γδ T-cells are effective killers of protozoan parasites. Sci Rep. 2014;4:5431.PubMedPubMedCentralCrossRef Peckham RK, Brill R, Foster DS, Bowen AL, Leigh JA, Coffey TJ, et al. Two distinct populations of bovine IL-17+ T-cells can be induced and WC1+IL-17+γδ T-cells are effective killers of protozoan parasites. Sci Rep. 2014;4:5431.PubMedPubMedCentralCrossRef
64.
go back to reference Saxena V, Mathur A, Krishnani N, Dhole TN. An insufficient anti-inflammatory cytokine response in mouse brain is associated with increased tissue pathology and viral load during Japanese encephalitis virus infection. Arch Virol. 2008;153(2):283–92.PubMedCrossRef Saxena V, Mathur A, Krishnani N, Dhole TN. An insufficient anti-inflammatory cytokine response in mouse brain is associated with increased tissue pathology and viral load during Japanese encephalitis virus infection. Arch Virol. 2008;153(2):283–92.PubMedCrossRef
65.
go back to reference Kulcsar KA, Baxter VK, Greene IP, Griffin DE. Interleukin 10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyelitis. Proc Natl Acd Sci USA. 2014;111(45):16053–8.CrossRef Kulcsar KA, Baxter VK, Greene IP, Griffin DE. Interleukin 10 modulation of pathogenic Th17 cells during fatal alphavirus encephalomyelitis. Proc Natl Acd Sci USA. 2014;111(45):16053–8.CrossRef
66.
go back to reference Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–8.PubMedCrossRef Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–8.PubMedCrossRef
67.
go back to reference Lochner M, Peduto L, Cherrier M, Sawa S, Langa F, Varona R, et al. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORgamma t+ T cells. J Exp Med. 2008;205(6):1381–93.PubMedPubMedCentralCrossRef Lochner M, Peduto L, Cherrier M, Sawa S, Langa F, Varona R, et al. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORgamma t+ T cells. J Exp Med. 2008;205(6):1381–93.PubMedPubMedCentralCrossRef
68.
go back to reference Van Der Ryst E. Maraviroc—a CCR5 antagonist for the treatment of HIV-1 infection. Front Immunol. 2015;6:277. Van Der Ryst E. Maraviroc—a CCR5 antagonist for the treatment of HIV-1 infection. Front Immunol. 2015;6:277.
Metadata
Title
CCR5 ameliorates Japanese encephalitis via dictating the equilibrium of regulatory CD4+Foxp3+ T and IL-17+CD4+ Th17 cells
Authors
Jin Hyoung Kim
Ajit Mahadev Patil
Jin Young Choi
Seong Bum Kim
Erdenebelig Uyangaa
Ferdaus Mohd Altaf Hossain
Sang-Youel Park
John Hwa Lee
Seong Kug Eo
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/s12974-016-0656-x

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue