404
Views
0
CrossRef citations to date
0
Altmetric
Review

Role of CXCL10 in central nervous system inflammation

&
Pages 1-18 | Published online: 27 Feb 2014

References

  • Eriksson P, Perfilieva E, Bjork-Eriksson T, et al. Neurogenesis in the adult human hippocampus. Nat Med. 1998;4(11):1313–1317.
  • van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, Gage FH. Functional neurogenesis in the adult hippocampus. Nature. 2002; 415(6875):1030–1034.
  • Galea I, Bechmann I, Perry VH. What is immune privilege (not)? Trends Immunol. 2007;28(1):12–18.
  • Ransohoff RM, Engelhardt B. The anatomical and cellular basis of immune surveillance in the central nervous system. Nat Rev Immunol. 2012;12(9):623–635.
  • Abbott NJ. Astrocyte-endothelial interactions and blood-brain barrier permeability. J Anat. 2002;200(6):629–638.
  • Ransohoff RM. The chemokine system in neuroinflammation: an update. J Infect Dis. 2002;186 Suppl 2:S152–S156.
  • Zlotnik A, Yoshie O. Chemokines: a new classification review system and their role in immunity. Immunity. 2000;12:121–127.
  • Vanguri P. Interferon-gamma-inducible genes in primary glial cells of the central nervous system: comparisons of astrocytes with microglia and Lewis with brown Norway rats. J Neuroimmunol. 1995;56(1):35–43.
  • Cole KE, Strick CA, Paradis TJ, et al. Interferon-inducible T cell alpha chemoattractant (I-TAC): a novel non-ELR CXC chemokine with potent activity on activated T cells through selective high affinity binding to CXCR3. J Exp Med. 1998;187(12):2009–2021.
  • Nomiyama H, Osada N, Yoshie O. The evolution of mammalian chemokine genes. Cytokine Growth Factor Rev. 2010;21(4):253–262.
  • Patel L, Charlton SJ, Chambers JK, Macphee CH. Expression and functional analysis of chemokine receptors in human peripheral blood leukocyte populations. Cytokine. 2001;14(1):27–36.
  • Belperio JA, Keane MP, Arenberg DA, et al. CXC chemokines in angiogenesis. J Leukoc Biol. 2000;68(1):1–8.
  • Doitsidou M, Reichman-Fried M, Stebler J, et al. Guidance of primordial germ cell migration by the chemokine SDF-1. Cell. 2002;111(5):647–659.
  • Zlotnik A, Yoshie O, Nomiyama H. The chemokine and chemokine receptor superfamilies and their molecular evolution. Genome Biol. 2006;7(12):243.
  • Luster AD, Jhanwar SC, Chaganti RS, Kersey JH, Ravetch JV. Interferon-inducible gene maps to a chromosomal band associated with a (4;11) translocation in acute leukemia cells. Proc Natl Acad Sci U S A. 1987;84(9):2868–2871.
  • Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature. 1985;315(6021):672–676.
  • Majumder SS, Zhou LZ, Chaturvedi PP, Babcock GG, Aras SS, Ransohoff RM. p48/STAT-1alpha-containing complexes play a predominant role in induction of IFN-gamma-inducible protein, 10 kDa (IP-10) by IFN-gamma alone or in synergy with TNF-alpha. J Immunol. 1998;161(9):4736–4744.
  • Ohmori Y, Hamilton TA. Cooperative interaction between interferon (IFN) stimulus response element and kappa B sequence motifs controls IFN gamma-and lipopolysaccharide-stimulated transcription from the murine IP-10 promoter. J Biol Chem. 1993;268(9):6677–6688.
  • Farber JM. Mig and IP-10: CXC chemokines that target lymphocytes. J Leukoc Biol. 1997;61(3):246–257.
  • Jensen S, Thomsen AR. Sensing of RNA viruses: a review of innate immune receptors involved in recognizing RNA virus invasion. J Virol. 2012;86(6):2900–2910.
  • Medoff BD, Wain JC, Seung E, et al. CXCR3 and its ligands in a murine model of obliterative bronchiolitis: regulation and function. J Immunol. 2006;176(11):7087–7095.
  • Hancock WW, Gao W, Csizmadia V, Faia KL, Shemmeri N, Luster AD. Donor-derived IP-10 initiates development of acute allograft rejection. J Exp Med. 2001;193(8):975–980.
  • Gasper NA, Petty CC, Schrum LW, Marriott I, Bost KL. Bacterium-induced CXCL10 secretion by osteoblasts can be mediated in part through toll-like receptor 4. Infect Immun. 2002;70(8):4075–4082.
  • Luster AD, Ravetch JV. Biochemical characterization of a gamma interferon-inducible cytokine (IP-10). J Exp Med. 1987;166(4):1084–1097.
  • McKimmie CS, Graham GJ. Astrocytes modulate the chemokine network in a pathogen-specific manner. Biochem Biophys Res Commun. 2010;394(4):1006–1011.
  • Xia MQ, Bacskai BJ, Knowles RB, Qin SX, Hyman BT. Expression of the chemokine receptor CXCR3 on neurons and the elevated expression of its ligand IP-10 in reactive astrocytes: in vitro ERK1/2 activation and role in Alzheimer’s disease. J Neuroimmunol. 2000;108(1–2):227–235.
  • Balashov KE, Rottman JB, Weiner HL, Hancock WW. CCR5(+) and CXCR3(+) T cells are increased in multiple sclerosis and their ligands MIP-1alpha and IP-10 are expressed in demyelinating brain lesions. Proc Natl Acad Sci U S A. 1999;96(12):6873–6878.
  • Phares TW, Stohlman SA, Hinton DR, Bergmann CC. Astrocyte-derived CXCL10 drives accumulation of antibody-secreting cells in the central nervous system during viral encephalomyelitis. J Virol. 2013;87(6):3382–3392.
  • Sørensen TL, Trebst C, Kivisäkk P, et al. Multiple sclerosis: a study of CXCL10 and CXCR3 co-localization in the inflamed central nervous system. J Neuroimmunol. 2002;127(1–2):59–68.
  • McKimmie CS, Roy D, Forster T, Fazakerley JK. Innate immune response gene expression profiles of N9 microglia are pathogen-type specific. J Neuroimmunol. 2006;175(1–2):128–141.
  • Cheeran MC, Hu S, Sheng WS, Peterson PK, Lokensgard JR. CXCL10 production from cytomegalovirus-stimulated microglia is regulated by both human and viral interleukin-10. J Virol. 2003;77(8):4502–4515.
  • Klein RS, Lin E, Zhang B, et al. Neuronal CXCL10 directs CD8+ T-cell recruitment and control of West Nile virus encephalitis. J Virol. 2005;79(17):11457–11466.
  • Campanella GSV, Tager AM, El Khoury JK, et al. Chemokine receptor CXCR3 and its ligands CXCL9 and CXCL10 are required for the development of murine cerebral malaria. Proc Natl Acad Sci U S A. 2008;105(12):4814–4819.
  • Loetscher M, Loetscher P, Brass N, Meese E, Moser B. Lymphocyte-specific chemokine receptor CXCR3: regulation, chemokine binding and gene localization. Eur J Immunol. 1998;28(11):3696–3705.
  • Qin S, Rottman JB, Myers P, et al. The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. J Clin Invest. 1998;101(4):746–754.
  • Loetscher P, Pellegrino A, Gong JH, et al. The ligands of CXC chemokine receptor 3, I-TAC, Mig, and IP10, are natural antagonists for CCR3. J Biol Chem. 2001;276(5):2986–2991.
  • Cox MA, Jenh CH, Gonsiorek W, et al. Human interferon-inducible 10-kDa protein and human interferon-inducible T cell alpha chemoattractant are allotopic ligands for human CXCR3: differential binding to receptor states. Mol Pharmacol. 2001;59(4):707–715.
  • Colvin RA, Campanella GSV, Manice LA, Luster AD. CXCR3 requires tyrosine sulfation for ligand binding and a second extracellular loop arginine residue for ligand-induced chemotaxis. Mol Cell Biol. 2006;26(15):5838–5849.
  • Strieter RM, Kunkel SL, Arenberg DA, Burdick MD, Polverini PJ. Interferon gamma-inducible protein 10 (IP-10), a member of the C-X-C chemokine family, is an inhibitor of angiogenesis. Biochem Biophys Res Commun. 1995;210(1):51–57.
  • Angiolillo AL, Sgadari C, Taub DD, et al. Human interferon-inducible protein 10 is a potent inhibitor of angiogenesis in vivo. J Exp Med. 1995;182:155–162.
  • Cole AM, Ganz TT, Liese AM, Burdick MD, Liu LL, Strieter RM. Cutting edge: IFN-inducible ELR- CXC chemokines display defensin-like antimicrobial activity. J Immunol. 2001;167(2):623–627.
  • Pertl U, Luster AD, Varki NM, et al. IFN-gamma-inducible protein-10 is essential for the generation of a protective tumor-specific CD8 T cell response induced by single-chain IL-12 gene therapy. J Immunol. 2001;166(11):6944–6951.
  • Groom JR, Richmond J, Murooka TT, et al. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity. 2012;37(6):1091–1103.
  • Sung JH, Zhang H, Moseman EA, et al. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell. 2012;150(6):1249–1263.
  • Christensen JE, Nansen A, Moos T, et al. Efficient T-cell surveillance of the CNS requires expression of the CXC chemokine receptor 3. J Neurosci. 2004;24(20):4849–4858.
  • Yates CC, Whaley D, Kulasekeran P, et al. Delayed and deficient dermal maturation in mice lacking the CXCR3 ELR-negative CXC chemokine receptor. Am J Pathol. 2007;171(2):484–495.
  • Groom JR, Luster AD. CXCR3 ligands: redundant, collaborative and antagonistic functions. Immunol Cell Biol. 2011;89(2):207–215.
  • Jacobs AH, Tavitian B. Noninvasive molecular imaging of neuroinflammation. J Cereb Blood Flow Metab. 2012;32(7):1393–1415.
  • Rivest S. Regulation of innate immune responses in the brain. Nat Rev Immunol. 2009;9(6):429–439.
  • Sotelo J, Corona T. Varicella zoster virus and relapsing remitting multiple sclerosis. Mult Scler Int. 2011;2011(3):1–5.
  • Lunemann JD, Kamradt T, Martin R, Munz C. Epstein-Barr virus: environmental trigger of multiple sclerosis? J Virol. 2007;81(13):6777–6784.
  • Simmons A. Herpesvirus and multiple sclerosis. Herpes. 2001;8(3):60–63.
  • Mostashari F, Bunning ML, Kitsutani PT, et al. Epidemic West Nile encephalitis, New York, 1999: results of a household-based seroepidemiological survey. Lancet. 2001;358(9278):261–264.
  • Gomez-Isla T, Price JL, McKeel DW, Morris JC, Growdon JH, Hyman BT. Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer’s disease. J Neurosci. 1996;16(14):4491–4500.
  • Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell. 2012;148(6):1204–1222.
  • Palop JJ, Mucke L. Amyloid-beta-induced neuronal dysfunction in Alzheimer’s disease: from synapses toward neural networks. Nat Neurosci. 2010;13(7):812–818.
  • Saruhan-Direskeneli G, Yentur SP, Akman-Demir G, Isik N, Serdaroglu P. Cytokines and chemokines in neuro-Behcet’s disease compared with multiple sclerosis and other neurological diseases. J Neuroimmunol. 2003;145(1–2):127–134.
  • Sui Y, Stehno-Bittel L, Li S, et al. CXCL10-induced cell death in neurons: role of calcium dysregulation. Eur J Neurosci. 2006;23(4):957–964.
  • Cho J, Nelson TE, Bajova H, Gruol DL. Chronic CXCL10 alters neuronal properties in rat hippocampal culture. J Neuroimmunol. 2009;207(1–2):92–100.
  • Nelson TE, Gruol DL. The chemokine CXCL10 modulates excitatory activity and intracellular calcium signaling in cultured hippocampal neurons. J Neuroimmunol. 2004;156(1–2):74–87.
  • Jucker M, Walker LC. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature. 2013;501(7465):45–51.
  • Head MW, Bunn TJR, Bishop MT, et al. Prion protein heterogeneity in sporadic but not variant Creutzfeldt-Jakob disease: UK cases 1991–2002. Ann Neurol. 2004;55(6):851–859.
  • Brown AR, Rebus S, McKimmie CS, Robertson K, Williams A, Fazakerley JK. Gene expression profiling of the preclinical scrapie-infected hippocampus. Biochem Biophys Res Commun. 2005;334(1):86–95.
  • Tribouillard-Tanvier D, Striebel JF, Peterson KE, Chesebro B. Analysis of protein levels of 24 cytokines in scrapie agent-infected brain and glial cell cultures from mice differing in prion protein expression levels. J Virol. 2009;83(21):11244–11253.
  • Riemer C, Schultz J, Burwinkel M, et al. Accelerated prion replication in, but prolonged survival times of, prion-infected CXCR3-/- mice. J Virol. 2008;82(24):12464–12471.
  • Müller M, Carter S, Hofer MJ, Campbell IL. Review: the chemokine receptor CXCR3 and its ligands CXCL9, CXCL10 and CXCL11 in neuroimmunity – a tale of conflict and conundrum. Neuropathol Appl Neurobiol. 2010;36(5):368–387.
  • Hsieh M-F, Lai S-L, Chen J-P, et al. Both CXCR3 and CXCL10/IFN-inducible protein 10 are required for resistance to primary infection by dengue virus. J Immunol. 2006;177(3):1855–1863.
  • Christensen JE, de Lemos C, Moos T, Christensen JP, Thomsen AR. CXCL10 is the key ligand for CXCR3 on CD8+ effector T cells involved in immune surveillance of the lymphocytic choriomeningitis virus-infected central nervous system. J Immunol. 2006;176(7):4235–4243.
  • Lassmann H, Ransohoff RM. The CD4-Th1 model for multiple sclerosis: a critical [correction of crucial] re-appraisal. Trends Immunol. 2004;25(3):132–137.
  • Sorensen TL, Tani M, Jensen J, et al. Expression of specific chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. J Clin Invest. 1999;103(6):807–815.
  • Kivisakk P, Trebst C, Liu Z, et al. T-cells in the cerebrospinal fluid express a similar repertoire of inflammatory chemokine receptors in the absence or presence of CNS inflammation: implications for CNS trafficking. Clin Exp Immunol. 2002;129(3):510–518.
  • El-behi M, Rostami A, Ciric B. Current views on the roles of Th1 and Th17 cells in experimental autoimmune encephalomyelitis. J Neuroimmune Pharmacol. 2010;5(2):189–197.
  • Franciotta D, Martino G, Zardini E, et al. Serum and CSF levels of MCP-1 and IP-10 in multiple sclerosis patients with acute and stable disease and undergoing immunomodulatory therapies. J Neuroimmunol. 2001;115(1–2):192–198.
  • Simpson JE, Newcombe J, Cuzner ML, Woodroofe MN. Expression of the interferon-gamma-inducible chemokines IP-10 and Mig and their receptor, CXCR3, in multiple sclerosis lesions. Neuropathol Appl Neurobiol. 2000;26(2):133–142.
  • Biddison WE, Cruikshank WW, Center DM, Pelfrey CM, Taub DD, Turner RV. CD8+ myelin peptide-specific T cells can chemoattract CD4+ myelin peptide-specific T cells: importance of IFN-inducible protein 10. J Immunol. 1998;160(1):444–448.
  • Gold R, Linington C, Lassmann H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain. 2006;129(8):1953–1971.
  • Sriram S, Steiner I. Experimental allergic encephalomyelitis: a misleading model of multiple sclerosis. Ann Neurol. 2005;58(6):939–945.
  • Ransohoff RM, Hamilton TA, Tani M, et al. Astrocyte expression of mRNA encoding cytokines IP-10 and JE/MCP-1 in experimental autoimmune encephalomyelitis. FASEB J. 1993;7(6):592–600.
  • Fife BT, Kennedy KJ, Paniagua MC, et al. CXCL10 (IFN-gamma-inducible protein-10) control of encephalitogenic CD4+ T cell accumulation in the central nervous system during experimental autoimmune encephalomyelitis. J Immunol. 2001;166(12):7617–7624.
  • Glabinski AR, Tani M, Tuohy VK, Tuthill RJ, Ransohoff RM. Central nervous system chemokine mRNA accumulation follows initial leukocyte entry at the onset of acute murine experimental autoimmune encephalomyelitis. Brain Behav Immun. 1995;9(4):315–330.
  • Wildbaum G, Netzer N, Karin N. Plasmid DNA encoding IFN-gamma-inducible protein 10 redirects antigen-specific T cell polarization and suppresses experimental autoimmune encephalomyelitis. J Immunol. 2002;168(11):5885–5892.
  • Narumi S, Kaburaki T, Yoneyama H, Iwamura H, Kobayashi Y, Matsushima K. Neutralization of IFN-inducible protein 10/CXCL10 exacerbates experimental autoimmune encephalomyelitis. Eur J Immunol. 2002;32(6):1784–1791.
  • Klein RS, Izikson L, Means T, et al. IFN-inducible protein 10/CXC chemokine ligand 10-independent induction of experimental autoimmune encephalomyelitis. J Immunol. 2004;172(1):550–559.
  • Lane TE, Asensio VC, Yu N, Paoletti AD, Campbell IL, Buchmeier MJ. Dynamic regulation of α-and β-chemokine expression in the central nervous system during mouse hepatitis virus-induced demyelinating disease. J Immunol. 1998;160(2):970–978.
  • Dufour JH, Dziejman M, Liu MT, Leung JH, Lane TE, Luster AD. IFN-gamma-inducible protein 10 (IP-10; CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking. J Immunol. 2002;168(7):3195–3204.
  • Stiles LN, Hosking MP, Edwards RA, Strieter RM, Lane TE. Differential roles for CXCR3 in CD4+ and CD8+ T cell trafficking following viral infection of the CNS. Eur J Immunol. 2006;36(3):613–622.
  • Liu MT, Armstrong DD, Hamilton TA, Lane TE. Expression of Mig (monokine induced by interferon-gamma) is important in T lymphocyte recruitment and host defense following viral infection of the central nervous system. J Immunol. 2001;166(3):1790–1795.
  • Christoffersen PJ, Volkert M, Rygaard J. Immunological unresponsiveness of nude mice to LCM virus infection. Acta Pathol Microbiol Scand C. 1976;84C(6):520–523.
  • Leist TP, Cobbold SP, Waldmann H, Aguet M, Zinkernagel RM. Functional analysis of T lymphocyte subsets in antiviral host defense. J Immunol. 1987;138(7):2278–2281.
  • Kim JV, Kang SS, Dustin ML, McGavern DB. Myelomonocytic cell recruitment causes fatal CNS vascular injury during acute viral meningitis. Nature. 2008;457(7226):191–195.
  • Christensen JE, Simonsen S, Fenger C, et al. Fulminant lymphocytic choriomeningitis virus-induced inflammation of the CNS involves a cytokine-chemokine-cytokine-chemokine cascade. J Immunol. 2009;182(2):1079–1087.
  • Asensio VC, Campbell IL. Chemokine gene expression in the brains of mice with lymphocytic choriomeningitis. J Virol. 1997;71(10):7832–7840.
  • Hofer MJ, Carter SL, Müller M, Campbell IL. Unaltered neurological disease and mortality in CXCR3-deficient mice infected intracranially with lymphocytic choriomeningitis Virus-Armstrong. Viral Immunol. 2008;21(4):425–433.
  • Fung-Leung WP, Kündig TM, Zinkernagel RM, Mak TW. Immune response against lymphocytic choriomeningitis virus infection in mice without CD8 expression. J Exp Med. 1991;174(6):1425–1429.
  • Andersen IH, Marker O, Thomsen AR. Breakdown of blood-brain barrier function in the murine lymphocytic choriomeningitis virus infection mediated by virus-specific CD8+ T cells. J Neuroimmunol. 1991;31(2):155–163.
  • Storm P, Bartholdy C, Sørensen MR, Christensen JP, Thomsen AR. Perforin-deficient CD8+ T cells mediate fatal lymphocytic choriomeningitis despite impaired cytokine production. J Virol. 2006;80(3):1222–1230.
  • Christen U, McGavern DB, Luster AD, von Herrath MG, Oldstone MB. Among CXCR3 chemokines, IFN-gamma-inducible protein of 10 kDa (CXC chemokine ligand (CXCL) 10) but not monokine induced by IFN-gamma (CXCL9) imprints a pattern for the subsequent development of autoimmune disease. J Immunol. 2003;171(12):6838–6845.
  • Wang Y, Lobigs M, Lee E, Müllbacher A. CD8+ T cells mediate recovery and immunopathology in West Nile virus encephalitis. J Virol. 2003;77(24):13323–13334.
  • 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):2641–2649.
  • Tsunoda I, Lane TE, Blackett J, Fujinami RS. Distinct roles for IP-10/CXCL10 in three animal models, Theiler’s virus infection, EAE, and MHV infection, for multiple sclerosis: implication of differing roles for IP-10. Mult Scler. 2004;10(1):26–34.
  • Theil DJ, Tsunoda I, Libbey JE, Derfuss TJ, Fujinami RS. Alterations in cytokine but not chemokine mRNA expression during three distinct Theiler’s virus infections. J Neuroimmunol. 2000;104(1):22–30.
  • Hoffman LM, Fife BT, Begolka WS, Miller SD, Karpus WJ. Central nervous system chemokine expression during Theiler’s virus-induced demyelinating disease. J Neurovirol. 1999;5(6):635–642.
  • Ransohoff RM, Wei T, Pavelko KD, Lee JC, Murray PD, Rodriguez M. Chemokine expression in the central nervous system of mice with a viral disease resembling multiple sclerosis: roles of CD4+ and CD8+ T cells and viral persistence. J Virol. 2002;76(5):2217–2224.
  • Ure DR, Lane TE, Liu MT, Rodriguez M. Neutralization of chemokines RANTES and MIG increases virus antigen expression and spinal cord pathology during Theiler’s virus infection. Int Immunol. 2005;17(5):569–579.
  • Wickham S, Lu B, Ash J, Carr DJ. Chemokine receptor deficiency is associated with increased chemokine expression in the peripheral and central nervous systems and increased resistance to herpetic encephalitis. J Neuroimmunol. 2005;162(1–2):51–59.
  • Carr DJ, Tomanek L. Herpes simplex virus and the chemokines that mediate the inflammation. Curr Top Microbiol Immunol. 2006;303:47–65.
  • Lokensgard JR, Hu S, Sheng W, et al. Robust expression of TNF-alpha, IL-1beta, RANTES, and IP-10 by human microglial cells during nonproductive infection with herpes simplex virus. J Neurovirol. 2001;7(3):208–219.
  • Sui Y, Potula R, Dhillon N, et al. Neuronal apoptosis is mediated by CXCL10 overexpression in simian human immunodeficiency virus encephalitis. Am J Pathol. 2004;164(5):1557–1566.
  • Wuest TR, Carr DJ. Dysregulation of CXCR3 signaling due to CXCL10 deficiency impairs the antiviral response to herpes simplex virus 1 infection. J Immunol. 2008;181(11):7985–7993.
  • Thapa M, Welner RS, Pelayo R, Carr DJ. CXCL9 and CXCL10 expression are critical for control of genital herpes simplex virus type 2 infection through mobilization of HSV-specific CTL and NK cells to the nervous system. J Immunol. 2008;180(2):1098–1106.
  • Hunt NH, Golenser J, Chan-Ling T, et al. Immunopathogenesis of cerebral malaria. Int J Parasitol. 2006;36(5):569–582.
  • Nitcheu J, Bonduelle O, Combadiere C, et al. Perforin-dependent brain-infiltrating cytotoxic CD8+ T lymphocytes mediate experimental cerebral malaria pathogenesis. J Immunol. 2003;170(4):2221–2228.
  • Hansen DS, Bernard NJ, Nie CQ, Schofield L. NK cells stimulate recruitment of CXCR3+ T cells to the brain during Plasmodium berghei-mediated cerebral malaria. J Immunol. 2007;178(9):5779–5788.
  • Van den Steen PE, Deroost K, Van Aelst I, et al. CXCR3 determines strain susceptibility to murine cerebral malaria by mediating T lymphocyte migration toward IFN-gamma-induced chemokines. Eur J Immunol. 2008;38(4):1082–1095.
  • Miu J, Mitchell AJ, Müller M, et al. Chemokine gene expression during fatal murine cerebral malaria and protection due to CXCR3 deficiency. J Immunol. 2008;180(2):1217–1230.
  • Armah HB, Wilson NO, Sarfo BY, et al. Cerebrospinal fluid and serum biomarkers of cerebral malaria mortality in Ghanaian children. Malar J. 2007;6:147.
  • Uzureau P, Uzureau S, Lecordier L, et al. Mechanism of Trypanosoma brucei gambiense resistance to human serum. Nature. 2013;501(7467):430–434.
  • Kennedy PG. Human African trypanosomiasis of the CNS: current issues and challenges. J Clin Invest. 2004;113(4):496–504.
  • Amin DN, Rottenberg ME, Thomsen AR, et al. Expression and role of CXCL10 during the encephalitic stage of experimental and clinical African trypanosomiasis. J Infect Dis. 2009;200(10):1556–1565.
  • Carter SL, Müller M, Manders PM, Campbell IL. Induction of the genes forCxcl9 andCxcl10 is dependent on IFN-γ but shows differential cellular expression in experimental autoimmune encephalomyelitis and by astrocytes and microgliain vitro. Glia. 2007;55(16):1728–1739.
  • Tiberti N, Matovu E, Hainard A, et al. New biomarkers for stage determination in Trypanosoma brucei rhodesiense sleeping sickness patients. Clin Transl Med. 2013;2(1):1.
  • Hainard A, Tiberti N, Robin X, et al. A combined CXCL10, CXCL8 and H-FABP panel for the staging of human African trypanosomiasis patients. PLoS Negl Trop Dis. 2009;3(6):e459.
  • Schall TJ, Proudfoot AE. Overcoming hurdles in developing successful drugs targeting chemokine receptors. Nat Rev Immunol. 2011;11(5):355–363.
  • Wijtmans M, Verzijl D, Leurs R, de Esch IJ, Smit MJ. Towards small-molecule CXCR3 ligands with clinical potential. Chem Med Chem. 2008;3(6):861–872.
  • Mohan K, Issekutz TB. Blockade of chemokine receptor CXCR3 inhibits T cell recruitment to inflamed joints and decreases the severity of adjuvant arthritis. J Immunol. 2007;179(12):8463–8469.
  • Gilliam BL, Riedel DJ, Redfield RR. Clinical use of CCR5 inhibitors in HIV and beyond. J Transl Med. 2011;9 Suppl 1:S9.
  • Gao P, Zhou X-Y, Yashiro-Ohtani Y, et al. The unique target specificity of a nonpeptide chemokine receptor antagonist: selective blockade of two Th1 chemokine receptors CCR5 and CXCR3. J Leukoc Biol. 2003;73(2):273–280.
  • Tonn GR, Wong SG, Wong SC, et al. An inhibitory metabolite leads to dose- and time-dependent pharmacokinetics of (R)-N- (3-(4-Ethoxy-phenyl)-4-oxo-3,4-dihydro-pyrido (2,3-d)pyrimidin-2-yl)-ethyl)-N-pyridin-3-yl-methyl-2-(4-trifluoromethoxy-phenyl)-acetamide (AMG 487) in human subjects after multiple dosing. Drug Metab Dispos. 2009;37(3):502–513.
  • Jopling LA, Watt GF, Fisher S, Birch H, Coggon S, Christie MI. Analysis of the pharmacokinetic/pharmacodynamic relationship of a small molecule CXCR3 antagonist, NBI-74330, using a murine CXCR3 internalization assay. Br J Pharmacol. 2007;152(8):1260–1271.
  • Horuk R. Chemokine receptor antagonists: overcoming developmental hurdles. Nat Rev Drug Discov. 2009;8(1):23–33.
  • Du X, Gustin DJ, Chen X, et al. Imidazo-pyrazine derivatives as potent CXCR3 antagonists. Bioorg Med Chem Lett. 2009;19(17):5200–5204.
  • Chen X, Mihalic J, Deignan J, et al. Discovery of potent and specific CXCR3 antagonists. Bioorg Med Chem Lett. 2012;22(1):357–362.
  • Campbell GL, Hills SL, Fischer M, et al. Estimated global incidence of Japanese encephalitis: a systematic review. Bull World Health Organ. 2011;89(10):766–774, 774A–774E.
  • Centers for Disease Control. West Nile virus disease and other arboviral diseases – United States, 2011. MMWR Morb Mortal Wkly Rep. 2012;61(27):510–514.
  • Samuel MA, Diamond MS. Pathogenesis of West Nile virus infection: a balance between virulence, innate and adaptive immunity, and viral evasion. J Virol. 2006;80(19):9349–9360.
  • Schnell MJ, McGettigan JP, Wirblich C, Papaneri A. The cell biology of rabies virus: using stealth to reach the brain. Nat Rev Micro. 2010;8(1):51–61.
  • Warrell M. Rabies and African bat lyssavirus encephalitis and its prevention. Int J Antimicrob Agents. 2010;36:S47–S52.
  • Sabah M, Mulcahy J, Zeman A. Herpes simplex encephalitis. BMJ. 2012;344:e3166.
  • Banatvala JE. Herpes simplex encephalitis. Lancet Infect Dis. 2011; 11(2):80–81.
  • Jain M, Duggal S, Chugh TD. Cytomegalovirus infection in non-immunosuppressed critically ill patients. J Infect Dev Ctries. 2011;5(8):571–579.
  • Cannon MJ, Schmid DS, Hyde TB. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol. 2010;20(4):202–213.
  • Obregon R, Chitnis K, Morry C, et al. Achieving polio eradication: a review of health communication evidence and lessons learned in India and Pakistan. Bull World Health Organ. 2009;87(8):624–630.
  • Gammelgaard LK, Colding H, Hartzen SH, Penkowa M. Meningococcal disease and future drug targets. CNS Neurol Disord Drug Targets. 2011;10(1):140–145.
  • Tzeng YL, Stephens DS. Epidemiology and pathogenesis of Neisseria meningitidis. Microbes Infect. 2000;2(6):687–700.
  • Fitch MT, van de Beek D. Emergency diagnosis and treatment of adult meningitis. Lancet Infect Dis. 2007;7(3):191–200.
  • Halperin JJ. Nervous system Lyme disease: is there a controversy? Semin Neurol. 2011;31(3):317–324.
  • O’Connell S. Lyme borreliosis: current issues in diagnosis and management. Curr Opin Infect Dis. 2010;23(3):231–235.
  • Idro R, Jenkins NE, Newton CR. Pathogenesis, clinical features, and neurological outcome of cerebral malaria. Lancet Neurol. 2005;4(12):827–840.
  • Shikani HJ, Freeman BD, Lisanti MP, Weiss LM, Tanowitz HB, Desruisseaux MS. Cerebral malaria: we have come a long way. Am J Pathol. 2012;181(5):1484–1492.
  • Kennedy PG. Diagnostic and neuropathogenesis issues in human African trypanosomiasis. Int J Parasitol. 2006;36(5):505–512.
  • Keita M, Bouteille B, Enanga B, Vallat JM, Dumas M. Trypanosoma brucei brucei: a long-term model of human African trypanosomiasis in mice, meningo-encephalitis, astrocytosis, and neurological disorders. Exp Parasitol. 1997;85(2):183–192.
  • Ross AG, McManus DP, Farrar J, Hunstman RJ, Gray DJ, Li Y-S. Neuroschistosomiasis. J Neurol. 2012;259(1):22–32.
  • Finsterer J, Auer H. Parasitoses of the human central nervous system. J Helminthol. 2013;87(3):257–270.
  • Feustel SM, Meissner M, Liesenfeld O. Toxoplasma gondii and the blood-brain barrier. Virulence. 2012;3(2):182–192.
  • Yan J, Huang B, Liu G, et al. Meta-analysis of prevention and treatment of toxoplasmic encephalitis in HIV-infected patients. Acta Trop. 2013;127(3):236–244.
  • Nie CQ, Bernard NJ, Norman MU, et al. IP-10-mediated T cell homing promotes cerebral inflammation over splenic immunity to malaria infection. PLoS Pathog. 2009;5(4):e1000369.
  • Khan IA, MacLean JA, Lee FS, et al. IP-10 is critical for effector T cell trafficking and host survival in Toxoplasma gondii infection. Immunity. 2000;12(5):483–494.
  • Lepej SZ, Rode OD, Jeren T, Vince A, Remenar A, Barsic B. Increased expression of CXCR3 and CCR5 on memory CD4+ T-cells migrating into the cerebrospinal fluid of patients with neuroborreliosis: the role of CXCL10 and CXCL11. J Neuroimmunol. 2005;163(1–2):128–134.
  • Giunti D, Borsellino G, Benelli R, et al. Phenotypic and functional analysis of T cells homing into the CSF of subjects with inflammatory diseases of the CNS. J Leukoc Biol. 2003;73(5):584–590.
  • Suzuki K, Kawauchi Y, Palaniyandi SS, et al. Blockade of interferon-gamma-inducible protein-10 attenuates chronic experimental colitis by blocking cellular trafficking and protecting intestinal epithelial cells. Pathol Int. 2007;57(7):413–420.
  • Yellin M, Paliienko I, Balanescu A, et al. A phase II, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of MDX-1100, a fully human anti-CXCL10 monoclonal antibody, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheum. 2012;64(6):1730–1739.
  • Baba M, Nishimura O, Kanzaki N, et al. A small-molecule, nonpeptide CCR5 antagonist with highly potent and selective anti-HIV-1 activity. Proc Natl Acad Sci U S A. 1999;96(10):5698–5703.
  • Johnson M, Li A-R, Liu J, et al. Discovery and optimization of a series of quinazolinone-derived antagonists of CXCR3. Bioorg Med Chem Lett. 2007;17(12):3339–3343.
  • Jenh C-H, Cox MA, Cui L, et al. A selective and potent CXCR3 antagonist SCH 546738 attenuates the development of autoimmune diseases and delays graft rejection. BMC Immunol. 2012;13(1):2.
  • Kim SH, Anilkumar GN, Zawacki LG, et al. III. Identification of novel CXCR3 chemokine receptor antagonists with a pyrazinyl-piperazinyl-piperidine scaffold. Bioorg Med Chem Lett. 2011;21(23):6982–6986.
  • Allen DR, Bolt A, Chapman GA, et al. Identification and structure-activity relationships of 1-aryl-3-piperidin-4-yl-urea derivatives as CXCR3 receptor antagonists. Bioorg Med Chem Lett. 2007;17(3):697–701.
  • Cole AG, Stroke IL, Brescia M-R, et al. Identification and initial evaluation of 4-N-aryl-[1,4]diazepane ureas as potent CXCR3 antagonists. Bioorg Med Chem Lett. 2006;16(1):200–203.
  • Stroke IL, Cole AG, Simhadri S, et al. Identification of CXCR3 receptor agonists in combinatorial small-molecule libraries. Biochem Biophys Res Commun. 2006;349(1):221–228.
  • Hayes ME, Wallace GA, Grongsaard P, et al. Discovery of small molecule benzimidazole antagonists of the chemokine receptor CXCR3. Bioorg Med Chem Lett. 2008;18(5):1573–1576.
  • Hayes ME, Breinlinger EC, Wallace GA, et al. Lead identification of 2-iminobenzimidazole antagonists of the chemokine receptor CXCR3. Bioorg Med Chem Lett. 2008;18(7):2414–2419.
  • Bongartz J-P, Buntinx M, Coesemans E, Hermans B, Lommen GV, Wauwe JV. Synthesis and structure-activity relationship of benzetimide derivatives as human CXCR3 antagonists. Bioorg Med Chem Lett. 2008;18(21):5819–5823.
  • Thoma G, Baenteli R, Lewis I, et al. Special ergolines efficiently inhibit the chemokine receptor CXCR3 in blood. Bioorg Med Chem Lett. 2011;21(16):4745–4749.
  • Thoma G, Baenteli R, Lewis I, et al. Special ergolines are highly selective, potent antagonists of the chemokine receptor CXCR3: discovery, characterization and preliminary SAR of a promising lead. Bioorg Med Chem Lett. 2009;19(21):6185–6188.