Abstract
Mouse hepatitis virus (MHV) is a positive strand RNA virus that causes an acute encephalomyelitis which later resolves into a chronic fulminating demyelinating disease. Cytokine production, chemokine secretion, and immune cell infiltration into the central nervous system are critical to control viral replication during acute infection. Despite potent anti – viral T lymphocyte activity, sterile immunity is not achieved, and MHV chronically persists within oligodendrocytes. Continued infiltration and activation of the immune system, a result of the lingering viral antigen and RNA within oligodendrocytes, lead directly to the development of an immune – mediated demyelination that bears remarkable similarities, both clinically and histologically, to the human demyelinating disease multiple sclerosis. MHV offers a unique model system for studying host defense during acute viral infection and immune – mediated demyelination during chronic infection.
Keywords: host defense, viral infection, multiple sclerosis, demyelination
I. Mouse Hepatitis Virus
MHV is a positive – strand RNA virus and a member of the family Coronaviridae, representing a significant ubiquitous group of viral pathogens that infect both humans and animals, causing respiratory, gastro-intestinal, and neurologic diseases. MHV, a group II coronavirus, is a natural pathogen of mice, normally infecting the liver, gastrointestinal tract, and central nervous system (CNS), causing a wide range of disease, including hepatitis, gastroenteritis, and acute and chronic encephalomyelitis1–3. MHV pathogenesis is dependent upon several factors including viral strain, mouse background, and inoculation route4. Structurally, MHV is comprised of three main proteins: the nucleocapsid (N, 60 kDa) which forms a helical complex with the genome, the membrane protein (M, 25 kDa) which associates with the nucleocapsid and aids in envelope formation and budding, and the extracellular spike glycoprotein (S, 180 kDa) which associates with the membrane protein and controls host cell receptor recognition and fusion2–4. MHV spike glycoprotein recognizes the host cell receptor carcinoembryonic antigen-cell adhesion molecule (CEACAM-1)5, 6 and dictates host pathogenesis and immune responses7–9.
Intracranial innoculation of susceptible strains of mice with neuroadapted strains of MHV induces an acute encephalomyelitis that evolves into a chronic fulminating demyelinating disease10. During acute encephalomyelitis, MHV infection stimulates the production of pro-inflammatory cytokines and chemokines that activate and attract the anti-viral arms of the immune system11–13. Anti-viral effector T lymphocytes are absolutely required for controlling viral replication via IFN-γ secretion or cytolytic activity14, 15. Eventually MHV is cleared below detectable levels; however sterile immunity is not achieved. The majority of mice that survive the initial acute infection develop an immune-mediated chronic demyelinating disease, characterized by viral persistence within white matter tracts of the spinal cord and continued T lymphocyte and macrophage infiltration16–19. Numerous clinical and histological similarities exist between MHV – induced demyelination and the human demyelinating disease multiple sclerosis (MS), making the MHV model system relevant for evaluating the underlying mechanisms associated with disease and repair. Moreover, given that the etiology of MS remains enigmatic20–23 and infectious agents such as viruses have been considered possible environmental triggering agents20, 24–27, the application of viral models of demyelination offers unique and important insight into the potential mechanisms that trigger and maintain immune – mediated demyelination. This review will provide a succinct overview of mechanisms associated both with host defense and disease progression in response to MHV infection of the CNS.
II. Acute MHV – induced encephalomyelitis
Following intracranial infection, MHV replicates first within the ependymal cells of the lateral ventricles before spreading throughout the parenchyma, targeting astrocytes, oligodendrocytes, and microglia28 (Figure 1). Neurons are spared within immunocompetent mice inoculated with neuroattenuated strains of MHV29–31. Following infection, MHV also traffics to the spinal cord, spreading through the cerebral spinal fluid and similarly infecting the local ependyma before disseminating throughout the parenchyma28. MHV infection of the CNS manifests significant early upregulation of inflammatory cytokines, chemokines, and matrix-metalloproteinases, all of which serve to initiate, attract, and support a robust host anti-viral response11–13, 32–37.
Figure 1. Mouse Hepatitis Virus Pathogenesis.
Following intracranial infection of susceptible mice, MHV replicates within astrocytes and oligodendrocytes. Among the earliest immune cells responding to infection, neutrophils are recruited into the CNS and serve to degrade the blood brain barrier through the release of matrix metalloproteinases (MMP), including MMP9, thus facilitating subsequent immune cell entry. Activated astrocytes secrete the T cell and macrophage chemoattractants CXCL9, CXCL10 and CCL5, directing virus-specific CD4+ and CD8+ T cells and macrophages into the CNS. Direct cytolytic activity by CD8+ T cells, mediated through perforin and granzyme B secretion, aids in viral clearance from infected astrocytes. Both CD4+ and CD8+ T cells also secrete IFNγ, activating macrophages and microglia, and promoting viral control within infected oligodendrocytes. Viral clearance from oligodendrocytes is incomplete and MHV antigen and/or RNA persist within oligodendrocytes. Chronic MHV persistence drives continued CXCL10/CCL5 secretion and T cell/macrophage infiltration within the CNS, leading to the development of immune – mediated demyelination. Activated macrophages/microglia present within the white matter digest myelin debris and serve to further enhance demyelination.
MHV infection of the CNS manifests significant early upregulation of inflammatory cytokines, chemokines, and matrix-metalloproteinases, all of which serve to initiate and support an anti-viral host response. Type I interferons (IFN-α and IFN-β), IL-1α, IL-1β, IL-6, IL-12, and TNFα are secreted following MHV infection12, 13, 32–34. Protective roles for the type I interferons during MHV infection have been well described. Exogenous treatment of either IFN-α or IFN-β limits MHV replication and dissemination within the CNS38, 39, while mice deficient in IFN-α/β-receptor quickly succumb to MHV infection40. The mechanisms of type I IFN in vivo protection are however complicated since MHV is resistant to IFN-β treatment in vitro41. Additionally, evidence suggests that MHV can shield their viral RNA genome from host pattern recognition receptors and therefore prevent IFN-β induction42, 43. Nevertheless, type I IFNs are clearly protective in vivo, and they may help to regulate innate and adaptive immune responses by enhancing MHC I expression44.
Neutrophils, NK cells, and macrophages are the primary innate immune cells recruited into the CNS immediately following MHV infection45, 46. Neutrophils are detectable within the CNS by day 2 post infection (p.i.) and peak at day 5 p.i.45, responding to chemotactic signals through the chemokine receptor CXCR2 (Hosking and Lane, unpublished observations). Neutrophils are primarily responsible for degrading the blood brain barrier (BBB) through matrix metalloproteinase (MMPs) secretion, facilitating extracellular matrix and basement lamina degradation and subsequent leukocyte migration47, 48 (Figure 1). Although neutrophils secrete MMP-936, 48, they are not the sole source of matrix metalloproteinases within the CNS, as MMP-3 and MMP-12 are derived from resident glia35. Nevertheless, neutrophils are indispensible for proper anti-viral responses, as their depletion prevents leukocyte entry into the CNS, thus limiting effective control of viral replication and spread48.
Similar to neutrophils, NK cells rapidly and transiently infiltrate into the CNS following MHV infection, peaking at day 5 p.i.46. Overexpression of viral derived CXCL10 in immune – deficient mice enhanced NK cell infiltration and reduced viral burden49, suggesting that NK cells may contribute in controlling viral replication. However, depletion of NK cells in immune – compromised mice did not enhance viral burden50, moreover the absence of NK cells within the CNS of immune – competent mice did not influence viral clearance or pathogenesis46, indicating that NK cells probably play little to no role in viral clearance under normal circumstances of MHV infection.
Inflammatory macrophages are first detectable by day 2 p.i.45, and unlike the transitory neutrophils and NK cells, they continue to accumulate within the CNS during the course of infection. Macrophage infiltration is dependent upon numerous chemokine signaling pathways including CCR2/CCL251, 52, CCL353, and CCL5/CCR554–56. Similar to NK cells, macrophages do not appear to perform any direct anti – viral activity within the CNS, as depletion of macrophages or neutralization of CCL5 during acute MHV infection does not enhance viral burden56, 57.
Both myeloid (CD11b+ CD11c+) and lymphoid (CD11b− CD11c+) derived dendritic cells (DC) are detectable within the CNS by day 2 p.i.58, though the chemotactic signals controlling their infiltration has not been fully explored. Migration of myeloid DCs to the draining lymph nodes is dependent, in part, on CCL3 expression58. Moreover, CCL3 deficiency reduces lymph node DC activation and skews TH1 anti-MHV responses58.
Early following MHV infection, virus – specific T cells are detectable within the local lymph nodes and spleen and subsequently migrate into the CNS59 (Figure 1). Protective immunity and anti-viral responses conform to a TH1 phenotype, broadly characterized by vigorous IFN-γ secretion and cytolytic activity14, 15, 60. Virus specific T cell generation is independent of IL-12 and/or IL-23, as viral clearance is unaffected following antibody neutralization of IL-23 and IL-12/2361 or genetic deletion of IL-1262. T cells isolated from the CNS are CXCR3 – reactive63 and their migration into the CNS is dependent upon the CXCR3 ligands CXCL9 and CXCL1063–66. Neutralization of CCL5 during infection also abrogates CD4+ and CD8+ T cell infiltration56, however CCR5 deficient CD8+ T cells adoptively transferred into MHV infected RAG1−/− recipients have no problem trafficking into the CNS67, while transferred CCR5 deficient CD4+ T cells do not efficiently enter the CNS68. Virus specific CD8+ T cells are the main cytolytic effector cell within the CNS and begin to accumulate by five days p.i.16, 59. CD8+ T cells are essential to controlling MHV replication56, 60; their accumulation within the CNS is concurrent with viral clearance from resident glia60, 69, 70. CD8+ T cells isolated from the CNS are cytolytic ex vivo70, 71, secreting IFN-γ and lytic molecules, including granzyme B and perforin17. In vivo, perforin – mediated cytolysis eliminates MHV from astrocytes and microglia14 and IFN-γ controls MHV replication within oligodendroglia15, 72. Recent evidence has also demonstrated that NKG2D signaling within the CNS enhances anti – viral CD8+ cytotoxic activity71.
Virus specific CD4+ T cells function in a supporting role for CD8+ T cells, and they are also critical in controlling MHV replication56, 73. In vivo CD4+ T cells secrete IFN-γ, facilitating viral clearance from oligodendroglia15, 72, upregulating MHC class II expression on microglia60 and MHC class I expression on oligodendoglia74, and thus enhancing immune cell activity within the CNS60, 75. CD8+ cytotoxicity and survival within the CNS is heavily dependent upon the presence of CD4+ T cells76, 77. How CD4+ T cells support and enhance CD8+ T cell activity is unknown, however it is assumed to be a secreted factor, since CD4+ T cells are spatially restricted near the vasculature, instead of migrating throughout the parenchyma like CD8+ T cells, possibly as a result of CD4+ T cell TIMP-1 expression35.
Antibody – secreting cells are detectable within the CNS of MHV infected mice by 5 days p.i., and neutralizing antibody is detectable within the serum by 10 days p.i.78. However, B cells do not participate in viral clearance during acute infection79, 80, rather MHV – specific antibodies prevent viral recrudescence in chronically infected mice79–81.
III. Chronic MHV – induced demyelination
After two weeks of MHV infection, viral loads with in the brain are reduced to below detectable levels by plaque assay. Sterile immunity is however not achieved, and viral antigen and/or RNA are detectable within oligodendrocytes up to a year p.i.11, 82 (Figure 1). Mechanisms contributing to viral persistence may include antigenic escape variants83 and generation of RNA quasispecies, although with regards to the later, the observed mutations are random and neither indicate specific immune pressure nor aid in escape from CD4+ or CD8+ surveillance84. More recently, CD8+ T cell exhaustion has been proposed to be a mechanism of MHV persistence. During chronic MHV infection, oligodendrocytes prominently express B7-H1 concurrently with infiltrating virus – specific CD8+ T cells that express PD-1. In the absence of B7-H1, MHV is cleared faster from the CNS, confirming that B7-H1/PD-1 signaling inhibits CD8+ anti-viral activity in vivo85.
Mice that survive acute MHV infection develop a chronic immune – mediated demyelinating disease. Infected mice first demonstrate signs of ascending demyelination during acute infection that range from limp tails to partial and complete hind limb paralysis. Analysis of the spinal cords of chronically – infected mice confirms that the loss of myelin integrity is associated with the continued presence of both viral antigen and inflammatory immune cells86 and not the apoptotic or necrotic death of myelinating oligodendrocytes87. No role for endogenous complement or antibody – mediated demyelination has been documented88, although exogenous auto-antibodies can exacerbate demyelination independent of complement during chronic infection89. Nevertheless, the immunopathology observed during chronic MHV infection resembles what is observed in the majority of active MS lesions10, 90, making chronic MHV infection an excellent model to study mechanisms of pathogenesis and potential treatments.
Concomitant with the absence of detectable infectious virus, total immune infiltration into the CNS wanes by two weeks p.i., yet virus – specific T cells and macrophages remain within the CNS for up to three months after infection16–19. Unlike in other models of CNS demyelination91–93 and in MS94–96, autoreactive T cells to defined myelin epitopes are not considered important in contributing to disease, indicating that chronic demyelination is mainly driven by anti – viral responses and not epitope spreading.
While both CD4+ and CD8+ T cells remain CXCR3+ during chronic infection63, only CD4+ T cells appear to rely upon CXCL10 for antiviral trafficking into the CNS; CD8+ T cell infiltration remains unaffected during CXCL10 neutralization19. Notably, CCL5 neutralization abrogates both CD4+ and CD8+ T cell accumulation during chronic infection55, indicating differential chemokine usage between the T cell subsets97.
The main effectors of demyelination during chronic MHV infection are T cells and macrophages (Figure 1). Both CD4+ and CD8+ T cells are important to the pathogenesis of chronic demyelination, although to differing degrees. Mice deficient in adaptive immune systems56, 73, 87 or CD4+ T cells 56 do not readily demyelinate, regardless of their ability to clear virus. Moreover, adoptive transfer of CD4+ T cells into infected RAG1 deficient hosts is sufficient to initiate demyelination56. CD4+ T cells also enhance demyelination, by attracting macrophages through CCL5 secretion56. Although it was reported that CD8−/− mice exhibit muted demyelination during chronic MHV infection56, IFN-γ dependent demyelination was observed following the transfer of CD8+ T cells into RAG-1−/− mice73, 98, 99, providing evidence that CD8+ T cells are capable of initiating and potentiating demyelination.
Although the exact mechanisms of demyelination have not be fully characterized, T lymphocyte secreted inflammatory cytokines including IFN-γ and TNF-α persist within the brain13 and/or spinal cord19, 71 up to four weeks p.i. even though infectious virus is no longer detectable. CD8+ cytolytic activity is muted during chronic infection, presumably as a result of decreasing viral antigen17, 70, however these cells still retain their capacity to secrete IFN-γ69.
Within chronically MHV infected mice, apoptosis has been observed associated with areas of pathological damage100. However no causal link between apoptosis and demyelination has been established, especially since RAG1−/− and wildtype mice display similar patterns of apoptosis, while only wildtype mice readily demyelinate87. Moreover, demyelination is observed during chronic MHV infection within mice that lack IFN-γR1 upon oligodendroglia, indicating that additional mechanisms for damage besides IFN-γ certainly exist.
Nevertheless, IFN-γ is directly harmful to both oligodendrocytes and oligodendrocyte precursor cells (OPC), reducing cell viability, inducing apoptosis, and in some cases necrosis101–109. IFN-γ can also indirectly induce microglia/macrophage secretion of TNF-α and nitric oxide, triggering oligodendrocyte cell death110, 111. Moreover, IFN-γ over expression during development results in widespread hypomyelination and oliogdendrocyte loss103, 112–114, while IFN-γ over expression abrogates remyelination and recovery during cuprizone – induced demyelination or peak EAE (experimental autoimmune encephalomyelitis) disease115. Within active MS lesions, IFN-γ is detectable by immunohistochemistry and is associated with oligodendrocyte apoptosis at the leading edges of the lesion104. Moreover, IFN-γ treatment of MS patients exacerbates disease116, whereas IFN-γ neutralization reduces disease disability117. Interestingly, within the spinal cords of chronically infected mice that have been treated with neutralizing antibodies for CXCL10, IFN-γ mRNA levels are reduced and this is associated with reduced demyelination and enhanced remyelination19.
As with other demyelinating diseases118, 119, ultrastructural analysis of MHV induced demyelinating lesions reveal myelin laden macrophages stripping and engulfing myelin120 (Figure 1). During chronic infection, macrophages are spatially associated within demyelinating white matter lesions of the spinal cord and are critical to demyelination. Neutralization of the potent macrophage chemokine CCL5 during chronic infection diminishes macrophage infiltration into the CNS and is associated with reduced demyelination55, 56. Moreover genetic silencing of CCR5, the chemokine receptor for CCL5, also prevents widespread demyelination, even in the absence of viral clearance54. Adoptive transfer of MHV – immunized splenocytes into RAG1−/− recipients resulted in the rapid demyelination, and this was associated with the widespread recruitment of activated macrophages to regions of pathology87. These observations are consistent with other models of demyelination, including, EAE121, 122 and cuprizone – induced demyelination123; likewise, reactive macrophages have also been described within demyelinating MS plaques124.
Although the main effectors of demyelination are certainly T cells and macrophages, this does not preclude the possibility that MHV may directly participate in damage, especially since oligodendrocytes are the main reservoir of MHV during chronic infection72, 125. In some MS lesions, oligodendrocyte apoptosis has also been observed126, 127, however the exact role of apoptosis in MS pathogenesis and pathology is unresolved128. In vitro, cultured murine oligodendrocytes are susceptible to MHV – induced apoptosis through FAS – spike glycoprotein interactions129–132. Moreover, the HIV protein Tat133 and the JC virus protein agnoprotein134 also enhance oligodendrocyte apoptosis in vitro. However, in vivo oligodendrocyte apoptosis during chronic MHV infection is not readily observed, and the presence of viral antigen does not appear to predispose an oligodendrocyte to apoptosis87. Therefore, it is likely that protective mechanisms exist during chronic infection that protect oligodendrocytes from MHV, IFN-γ, and other apoptotic inducers.
Within chronic MHV demyelinating lesions, endogenous remyelination has been observed135–137. Moreover, remyelination and actively proliferating oligodendrocytes have been observed within MS lesions, indicating that repair can occur concurrently with acute or chronic inflammatory events138, 139. In vitro, growth factors and cytokines including insulin – like growth factor (IGF), ciliary neurotrophic factor (CNTF), leukemia inhibitory factor (LIF), neurotrophin-3 (NT3), and platelet-derived growth factor (PDGF) promote oligodendrocyte survival140–143. Additionally, the cytokine IL-11 which has been detected on reactive astrocytes within MS lesions144 and in MHV infected astrocytes145 has recently been demonstrated to enhance oligodendrocyte survival in vitro144. Studies by Kilpatrick and colleagues146–148 have further demonstrated a potent role for LIF in limiting demyelination during EAE by enhancing oligodendrocyte survival in vivo. Taken together these data indicate the complex protective and damaging inflammatory environment that exists within demyelinating lesions.
IV. Conclusions
This review highlights MHV as a model system for viral – induced neurologic disease. Specifically, MHV offers a platform for differentially studying the underlying mechanisms that dictate host defense during acute viral infection and later contribute to demyelination during chronic viral persistence. Notably, the pathology observed during chronic MHV demyelination closely parallels the damage observed in MS patients. Recent documented inconsistencies between EAE and MS116, 149, 150, where protective treatments in EAE exacerbate or have no effect on MS patients, underscore the necessity for the broader application of diverse demyelinating models that can complement each other and lead to a greater understanding of the fundamental processes that lead to demyelination and the development of MS.
Acknowledgments
This work was supported by National Multiple Sclerosis Society grant 3278 and National Institutes of Health grant NS41249 to T.E.L. M.P.H. was supported by NIH T32 AI-060573.
References
- 1.Cheever FS, et al. A murine virus (JHM) causing disseminated encephalomyelitis with extensive destruction of myelin. Journal of Experimental Medicine. 1949;90(3):181–210. doi: 10.1084/jem.90.3.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Holmes K, Lai M. Coronaviridae: The Viruses and Their Replication. In: Fields BN, Knipe DM, Howley PM, editors. Fields Virology. Lippincott-Raven Publishers; Philadelphia: 1996. pp. 1075–1094. [Google Scholar]
- 3.McIntosh K. Coronaviruses. In: Fields BN, Knipe DM, Howley PM, editors. Fields Virology. Lippincott-Raven Publishers; Philadelphia: 1996. pp. 1095–1104. [Google Scholar]
- 4.Perlman SR, Lane TE, Buchmeier MJ. Coronaviruses: Hepatitis, peritonitis, and central nervous system disease. In: Cunningham MW, Fujinami RS, editors. Effects of Microbes on the Immune System. Lippincott Williams & Wilkins; Philadelphia: 1999. pp. 331–348. [Google Scholar]
- 5.Compton SR, et al. Coronavirus species specificity: murine coronavirus binds to a mouse-specific epitope on its carcinoembryonic antigen-related receptor glycoprotein. J Virol. 1992;66(12):7420–8. doi: 10.1128/jvi.66.12.7420-7428.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Williams RK, et al. Purification of the 110-kilodalton glycoprotein receptor for mouse hepatitis virus (MHV)-A59 from mouse liver and identification of a nonfunctional, homologous protein in MHV-resistant SJL/J mice. J Virol. 1990;64(8):3817–23. doi: 10.1128/jvi.64.8.3817-3823.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ontiveros E, et al. Enhanced virulence mediated by the murine coronavirus, mouse hepatitis virus strain JHM, is associated with a glycine at residue 310 of the spike glycoprotein. J Virol. 2003;77(19):10260–9. doi: 10.1128/JVI.77.19.10260-10269.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Iacono KT, Kazi L, Weiss SR. Both spike and background genes contribute to murine coronavirus neurovirulence. J Virol. 2006;80(14):6834–43. doi: 10.1128/JVI.00432-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Phillips JJ, et al. Murine coronavirus spike glycoprotein mediates degree of viral spread, inflammation, and virus-induced immunopathology in the central nervous system. Virology. 2002;301(1):109–20. doi: 10.1006/viro.2002.1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Houtman JJ, Fleming JO. Pathogenesis of mouse hepatitis virus-induced demyelination. J Neurovirol. 1996;2(6):361–76. doi: 10.3109/13550289609146902. [DOI] [PubMed] [Google Scholar]
- 11.Lane TE, et al. Dynamic regulation of alpha- and beta-chemokine expression in the central nervous system during mouse hepatitis virus-induced demyelinating disease. J Immunol. 1998;160(2):970–8. [PubMed] [Google Scholar]
- 12.Pearce BD, et al. Cytokine induction during T-cell-mediated clearance of mouse hepatitis virus from neurons in vivo. J Virol. 1994;68(9):5483–95. doi: 10.1128/jvi.68.9.5483-5495.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Parra B, et al. Kinetics of cytokine mRNA expression in the central nervous system following lethal and nonlethal coronavirus-induced acute encephalomyelitis. Virology. 1997;233(2):260–70. doi: 10.1006/viro.1997.8613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lin MT, Stohlman SA, Hinton DR. Mouse hepatitis virus is cleared from the central nervous systems of mice lacking perforin-mediated cytolysis. J Virol. 1997;71(1):383–91. doi: 10.1128/jvi.71.1.383-391.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Parra B, et al. IFN-gamma is required for viral clearance from central nervous system oligodendroglia. J Immunol. 1999;162(3):1641–7. [PubMed] [Google Scholar]
- 16.Marten NW, Stohlman SA, Bergmann CC. Role of viral persistence in retaining CD8(+) T cells within the central nervous system. J Virol. 2000;74(17):7903–10. doi: 10.1128/jvi.74.17.7903-7910.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ramakrishna C, et al. Differential regulation of primary and secondary CD8+ T cells in the central nervous system. J Immunol. 2004;173(10):6265–73. doi: 10.4049/jimmunol.173.10.6265. [DOI] [PubMed] [Google Scholar]
- 18.Castro RF, et al. Coronavirus-induced demyelination occurs in the presence of virus-specific cytotoxic T cells. Virology. 1994;200(2):733–43. doi: 10.1006/viro.1994.1237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Liu MT, Keirstead HS, Lane TE. Neutralization of the chemokine CXCL10 reduces inflammatory cell invasion and demyelination and improves neurological function in a viral model of multiple sclerosis. J Immunol. 2001;167(7):4091–7. doi: 10.4049/jimmunol.167.7.4091. [DOI] [PubMed] [Google Scholar]
- 20.Ascherio A, Munger KL. Environmental risk factors for multiple sclerosis. Part I: the role of infection. Ann Neurol. 2007;61(4):288–99. doi: 10.1002/ana.21117. [DOI] [PubMed] [Google Scholar]
- 21.Ascherio A, Munger KL. Environmental risk factors for multiple sclerosis. Part II: Noninfectious factors. Ann Neurol. 2007;61(6):504–13. doi: 10.1002/ana.21141. [DOI] [PubMed] [Google Scholar]
- 22.Hafler DA, et al. Risk alleles for multiple sclerosis identified by a genomewide study. N Engl J Med. 2007;357(9):851–62. doi: 10.1056/NEJMoa073493. [DOI] [PubMed] [Google Scholar]
- 23.Lincoln MR, et al. A predominant role for the HLA class II region in the association of the MHC region with multiple sclerosis. Nat Genet. 2005;37(10):1108–12. doi: 10.1038/ng1647. [DOI] [PubMed] [Google Scholar]
- 24.Buljevac D, et al. Prospective study on the relationship between infections and multiple sclerosis exacerbations. Brain. 2002;125(Pt 5):952–60. doi: 10.1093/brain/awf098. [DOI] [PubMed] [Google Scholar]
- 25.Panitch HS. Influence of infection on exacerbations of multiple sclerosis. Ann Neurol. 1994;36(Suppl):S25–8. doi: 10.1002/ana.410360709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Thacker EL, Mirzaei F, Ascherio A. Infectious mononucleosis and risk for multiple sclerosis: a meta-analysis. Ann Neurol. 2006;59(3):499–503. doi: 10.1002/ana.20820. [DOI] [PubMed] [Google Scholar]
- 27.Sotelo J, et al. Varicella-zoster virus in cerebrospinal fluid at relapses of multiple sclerosis. Ann Neurol. 2008;63(3):303–11. doi: 10.1002/ana.21316. [DOI] [PubMed] [Google Scholar]
- 28.Wang FI, et al. Sequential infection of glial cells by the murine hepatitis virus JHM strain (MHV-4) leads to a characteristic distribution of demyelination. Lab Invest. 1992;66(6):744–54. [PubMed] [Google Scholar]
- 29.Buchmeier MJ, et al. Murine hepatitis virus-4 (strain JHM)-induced neurologic disease is modulated in vivo by monoclonal antibody. Virology. 1984;132(2):261–70. doi: 10.1016/0042-6822(84)90033-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fleming JO, et al. Antigenic relationships of murine coronaviruses: analysis using monoclonal antibodies to JHM (MHV-4) virus. Virology. 1983;131(2):296–307. doi: 10.1016/0042-6822(83)90498-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ireland DD, et al. Type I interferons are essential in controlling neurotropic coronavirus infection irrespective of functional CD8 T cells. J Virol. 2008;82(1):300–10. doi: 10.1128/JVI.01794-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rempel JD, et al. Viral induction of central nervous system innate immune responses. J Virol. 2005;79(7):4369–81. doi: 10.1128/JVI.79.7.4369-4381.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Rempel JD, et al. Differential regulation of innate and adaptive immune responses in viral encephalitis. Virology. 2004;318(1):381–92. doi: 10.1016/j.virol.2003.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sun N, et al. Activation of astrocytes in the spinal cord of mice chronically infected with a neurotropic coronavirus. Virology. 1995;213(2):482–93. doi: 10.1006/viro.1995.0021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhou J, et al. Expression of matrix metalloproteinases and their tissue inhibitor during viral encephalitis. J Virol. 2005;79(8):4764–73. doi: 10.1128/JVI.79.8.4764-4773.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhou J, et al. Matrix metalloproteinase expression correlates with virulence following neurotropic mouse hepatitis virus infection. J Virol. 2002;76(15):7374–84. doi: 10.1128/JVI.76.15.7374-7384.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Glass WG, et al. Mouse hepatitis virus infection of the central nervous system: chemokine-mediated regulation of host defense and disease. Viral Immunol. 2002;15(2):261–72. doi: 10.1089/08828240260066215. [DOI] [PubMed] [Google Scholar]
- 38.Minagawa H, et al. Protective effect of recombinant murine interferon beta against mouse hepatitis virus infection. Antiviral Res. 1987;8(2):85–95. doi: 10.1016/0166-3542(87)90079-9. [DOI] [PubMed] [Google Scholar]
- 39.Smith AL, Barthold SW, Beck DS. Intranasally administered alpha/beta interferon prevents extension of mouse hepatitis virus, strain JHM, into the brains of BALB/cByJ mice. Antiviral Res. 1987;8(5–6):239–45. doi: 10.1016/S0166-3542(87)80002-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Cervantes-Barragan L, et al. Control of coronavirus infection through plasmacytoid dendritic cell-derived type I interferon. Blood. 2006 doi: 10.1182/blood-2006-05-023770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Roth-Cross JK, et al. Inhibition of the alpha/beta interferon response by mouse hepatitis virus at multiple levels. J Virol. 2007;81(13):7189–99. doi: 10.1128/JVI.00013-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Versteeg GA, et al. Group 2 coronaviruses prevent immediate early interferon induction by protection of viral RNA from host cell recognition. Virology. 2007;361(1):18–26. doi: 10.1016/j.virol.2007.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhou H, Perlman S. Mouse hepatitis virus does not induce Beta interferon synthesis and does not inhibit its induction by double-stranded RNA. J Virol. 2007;81(2):568–74. doi: 10.1128/JVI.01512-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Akwa Y, et al. Transgenic expression of IFN-alpha in the central nervous system of mice protects against lethal neurotropic viral infection but induces inflammation and neurodegeneration. J Immunol. 1998;161(9):5016–26. [PubMed] [Google Scholar]
- 45.Templeton SP, et al. Maturation and localization of macrophages and microglia during infection with a neurotropic murine coronavirus. Brain Pathol. 2008;18(1):40–51. doi: 10.1111/j.1750-3639.2007.00098.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zuo J, et al. Mouse hepatitis virus pathogenesis in the central nervous system is independent of IL-15 and natural killer cells. Virology. 2006;350(1):206–15. doi: 10.1016/j.virol.2006.01.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yong VW, et al. Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci. 2001;2(7):502–11. doi: 10.1038/35081571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Zhou J, et al. Neutrophils promote mononuclear cell infiltration during viral-induced encephalitis. J Immunol. 2003;170(6):3331–6. doi: 10.4049/jimmunol.170.6.3331. [DOI] [PubMed] [Google Scholar]
- 49.Trifilo MJ, et al. CXC chemokine ligand 10 controls viral infection in the central nervous system: evidence for a role in innate immune response through recruitment and activation of natural killer cells. J Virol. 2004;78(2):585–94. doi: 10.1128/JVI.78.2.585-594.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Muse M, et al. Insertion of the CXC chemokine ligand 9 (CXCL9) into the mouse hepatitis virus genome results in protection from viral-induced encephalitis and hepatitis. Virology. 2008;382(2):132–44. doi: 10.1016/j.virol.2008.09.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen BP, Kuziel WA, Lane TE. Lack of CCR2 results in increased mortality and impaired leukocyte activation and trafficking following infection of the central nervous system with a neurotropic coronavirus. J Immunol. 2001;167(8):4585–92. doi: 10.4049/jimmunol.167.8.4585. [DOI] [PubMed] [Google Scholar]
- 52.Held KS, et al. Differential roles of CCL2 and CCR2 in host defense to coronavirus infection. Virology. 2004;329(2):251–60. doi: 10.1016/j.virol.2004.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Trifilo MJ, et al. CC chemokine ligand 3 (CCL3) regulates CD8(+)-T-cell effector function and migration following viral infection. J Virol. 2003;77(7):4004–14. doi: 10.1128/JVI.77.7.4004-4014.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Glass WG, et al. Reduced macrophage infiltration and demyelination in mice lacking the chemokine receptor CCR5 following infection with a neurotropic coronavirus. Virology. 2001;288(1):8–17. doi: 10.1006/viro.2001.1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Glass WG, et al. Antibody targeting of the CC chemokine ligand 5 results in diminished leukocyte infiltration into the central nervous system and reduced neurologic disease in a viral model of multiple sclerosis. J Immunol. 2004;172(7):4018–25. doi: 10.4049/jimmunol.172.7.4018. [DOI] [PubMed] [Google Scholar]
- 56.Lane TE, 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. doi: 10.1128/jvi.74.3.1415-1424.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Xue S, et al. Depletion of blood-borne macrophages does not reduce demyelination in mice infected with a neurotropic coronavirus. J Virol. 1999;73(8):6327–34. doi: 10.1128/jvi.73.8.6327-6334.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Trifilo MJ, Lane TE. The CC chemokine ligand 3 regulates CD11c+CD11b+CD8alpha- dendritic cell maturation and activation following viral infection of the central nervous system: implications for a role in T cell activation. Virology. 2004;327(1):8–15. doi: 10.1016/j.virol.2004.06.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Marten NW, et al. Kinetics of virus-specific CD8+-T-cell expansion and trafficking following central nervous system infection. J Virol. 2003;77(4):2775–8. doi: 10.1128/JVI.77.4.2775-2778.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bergmann CC, et al. Perforin-mediated effector function within the central nervous system requires IFN-gamma-mediated MHC up-regulation. J Immunol. 2003;170(6):3204–13. doi: 10.4049/jimmunol.170.6.3204. [DOI] [PubMed] [Google Scholar]
- 61.Held KS, et al. Generation of a Protective T-Cell Response Following Coronavirus Infection of the Central Nervous System Is Not Dependent on IL-12/23 Signaling. Viral Immunol. 2008;21(2):173–88. doi: 10.1089/vim.2008.0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kapil P, et al. Interleukin-12 (IL-12), but not IL-23, deficiency ameliorates viral encephalitis without affecting viral control. J Virol. 2009;83(12):5978–86. doi: 10.1128/JVI.00315-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Stiles LN, et al. Differential roles for CXCR3 in CD4+ and CD8+ T cell trafficking following viral infection of the CNS. Eur J Immunol. 2006;36(3):613–22. doi: 10.1002/eji.200535509. [DOI] [PubMed] [Google Scholar]
- 64.Liu MT, et al. 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):1790–5. doi: 10.4049/jimmunol.166.3.1790. [DOI] [PubMed] [Google Scholar]
- 65.Liu MT, et al. The T cell chemoattractant IFN-inducible protein 10 is essential in host defense against viral-induced neurologic disease. J Immunol. 2000;165(5):2327–30. doi: 10.4049/jimmunol.165.5.2327. [DOI] [PubMed] [Google Scholar]
- 66.Walsh KB, et al. Expression of CXC chemokine ligand 10 from the mouse hepatitis virus genome results in protection from viral-induced neurological and liver disease. J Immunol. 2007;179(2):1155–65. doi: 10.4049/jimmunol.179.2.1155. [DOI] [PubMed] [Google Scholar]
- 67.Glass WG, Lane TE. Functional analysis of the CC chemokine receptor 5 (CCR5) on virus-specific CD8+ T cells following coronavirus infection of the central nervous system. Virology. 2003;312(2):407–14. doi: 10.1016/S0042-6822(03)00237-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Glass WG, Lane TE. Functional expression of chemokine receptor CCR5 on CD4(+) T cells during virus-induced central nervous system disease. J Virol. 2003;77(1):191–8. doi: 10.1128/JVI.77.1.191-198.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bergmann CC, Lane TE, Stohlman SA. Coronavirus infection of the central nervous system: host-virus stand-off. Nat Rev Microbiol. 2006;4(2):121–32. doi: 10.1038/nrmicro1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Bergmann CC, et al. Inverted immunodominance and impaired cytolytic function of CD8+ T cells during viral persistence in the central nervous system. J Immunol. 1999;163(6):3379–87. [PubMed] [Google Scholar]
- 71.Walsh KB, Lanier LL, Lane TE. NKG2D receptor signaling enhances cytolytic activity by virus-specific CD8+ T cells: evidence for a protective role in virus-induced encephalitis. J Virol. 2008;82(6):3031–44. doi: 10.1128/JVI.02033-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Gonzalez JM, et al. Inhibition of interferon-gamma signaling in oligodendroglia delays coronavirus clearance without altering demyelination. Am J Pathol. 2006;168(3):796–804. doi: 10.2353/ajpath.2006.050496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wu GF, et al. CD4 and CD8 T cells have redundant but not identical roles in virus-induced demyelination. J Immunol. 2000;165(4):2278–86. doi: 10.4049/jimmunol.165.4.2278. [DOI] [PubMed] [Google Scholar]
- 74.Malone KE, et al. Induction of class I antigen processing components in oligodendroglia and microglia during viral encephalomyelitis. Glia. 2008;56(4):426–35. doi: 10.1002/glia.20625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bergmann CC, et al. Perforin and gamma interferon-mediated control of coronavirus central nervous system infection by CD8 T cells in the absence of CD4 T cells. J Virol. 2004;78(4):1739–50. doi: 10.1128/JVI.78.4.1739-1750.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Stohlman SA, et al. CTL effector function within the central nervous system requires CD4+ T cells. J Immunol. 1998;160(6):2896–904. [PubMed] [Google Scholar]
- 77.Zhou J, et al. Maintenance of CD8+ T cells during acute viral infection of the central nervous system requires CD4+ T cells but not interleukin-2. Viral Immunol. 2005;18(1):162–9. doi: 10.1089/vim.2005.18.162. [DOI] [PubMed] [Google Scholar]
- 78.Tschen SI, et al. Recruitment kinetics and composition of antibody-secreting cells within the central nervous system following viral encephalomyelitis. J Immunol. 2002;168(6):2922–9. doi: 10.4049/jimmunol.168.6.2922. [DOI] [PubMed] [Google Scholar]
- 79.Ramakrishna C, et al. Control of central nervous system viral persistence by neutralizing antibody. J Virol. 2003;77(8):4670–8. doi: 10.1128/JVI.77.8.4670-4678.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ramakrishna C, et al. Mechanisms of central nervous system viral persistence: the critical role of antibody and B cells. J Immunol. 2002;168(3):1204–11. doi: 10.4049/jimmunol.168.3.1204. [DOI] [PubMed] [Google Scholar]
- 81.Lin MT, et al. Antibody prevents virus reactivation within the central nervous system. J Immunol. 1999;162(12):7358–68. [PubMed] [Google Scholar]
- 82.Adami C, et al. Evolution of mouse hepatitis virus (MHV) during chronic infection: quasispecies nature of the persisting MHV RNA. Virology. 1995;209(2):337–46. doi: 10.1006/viro.1995.1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Pewe L, et al. Cytotoxic T cell-resistant variants are selected in a virus-induced demyelinating disease. Immunity. 1996;5(3):253–62. doi: 10.1016/s1074-7613(00)80320-9. [DOI] [PubMed] [Google Scholar]
- 84.Bergmann C, et al. Variability of persisting MHV RNA sequences constituting immune and replication-relevant domains. Virology. 1998;244(2):563–72. doi: 10.1006/viro.1998.9147. [DOI] [PubMed] [Google Scholar]
- 85.Phares TW, et al. Target-dependent B7-H1 regulation contributes to clearance of central nervous system infection and dampens morbidity. J Immunol. 2009;182(9):5430–8. doi: 10.4049/jimmunol.0803557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Stohlman SA, Hinton David R. Viral Induced Demyelination. Brain Pathology. 2001;11(1):92–106. doi: 10.1111/j.1750-3639.2001.tb00384.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wu GF, Perlman S. Macrophage infiltration, but not apoptosis, is correlated with immune-mediated demyelination following murine infection with a neurotropic coronavirus. J Virol. 1999;73(10):8771–80. doi: 10.1128/jvi.73.10.8771-8780.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Matthews AE, et al. Neither B cells nor T cells are required for CNS demyelination in mice persistently infected with MHV-A59. J Neurovirol. 2002;8(3):257–64. doi: 10.1080/13550280290049697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Burrer R, et al. Exacerbated pathology of viral encephalitis in mice with central nervous system-specific autoantibodies. Am J Pathol. 2007;170(2):557–66. doi: 10.2353/ajpath.2007.060893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Matthews AE, Weiss SR, Paterson Y. Murine hepatitis virus--a model for virus-induced CNS demyelination. J Neurovirol. 2002;8(2):76–85. doi: 10.1080/13550280290049534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Miller SD, et al. Persistent infection with Theiler’s virus leads to CNS autoimmunity via epitope spreading. Nat Med. 1997;3(10):1133–6. doi: 10.1038/nm1097-1133. [DOI] [PubMed] [Google Scholar]
- 92.Katz-Levy Y, et al. Temporal development of autoreactive Th1 responses and endogenous presentation of self myelin epitopes by central nervous system-resident APCs in Theiler’s virus-infected mice. J Immunol. 2000;165(9):5304–14. doi: 10.4049/jimmunol.165.9.5304. [DOI] [PubMed] [Google Scholar]
- 93.McMahon EJ, et al. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat Med. 2005;11(3):335–9. doi: 10.1038/nm1202. [DOI] [PubMed] [Google Scholar]
- 94.Tuohy VK, et al. Diversity and plasticity of self recognition during the development of multiple sclerosis. J Clin Invest. 1997;99(7):1682–90. doi: 10.1172/JCI119331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Goebels N, et al. Repertoire dynamics of autoreactive T cells in multiple sclerosis patients and healthy subjects: epitope spreading versus clonal persistence. Brain. 2000;123(Pt 3):508–18. doi: 10.1093/brain/123.3.508. [DOI] [PubMed] [Google Scholar]
- 96.Tuohy VK, et al. Spontaneous regression of primary autoreactivity during chronic progression of experimental autoimmune encephalomyelitis and multiple sclerosis. J Exp Med. 1999;189(7):1033–42. doi: 10.1084/jem.189.7.1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Stiles LN, Liu MT, Kane JAC, Lane TE. CXCL10 and trafficking of virus-specific T cells during coronavirus-induced demyelination. Autoimmunity. 2009 doi: 10.1080/08916930902810708. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Pewe L, Haring J, Perlman S. CD4 T-cell-mediated demyelination is increased in the absence of gamma interferon in mice infected with mouse hepatitis virus. J Virol. 2002;76(14):7329–33. doi: 10.1128/JVI.76.14.7329-7333.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Pewe L, Perlman S. Cutting edge: CD8 T cell-mediated demyelination is IFN-gamma dependent in mice infected with a neurotropic coronavirus. J Immunol. 2002;168(4):1547–51. doi: 10.4049/jimmunol.168.4.1547. [DOI] [PubMed] [Google Scholar]
- 100.Schwartz T, Fu L, Lavi E. Differential induction of apoptosis in demyelinating and nondemyelinating infection by mouse hepatitis virus. J Neurovirol. 2002;8(5):392–9. doi: 10.1080/13550280260422695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Loughlin AJ, et al. Myelination and remyelination of aggregate rat brain cell cultures enriched with macrophages. J Neurosci Res. 1997;47(4):384–92. [PubMed] [Google Scholar]
- 102.Baerwald KD, Popko B. Developing and mature oligodendrocytes respond differently to the immune cytokine interferon-gamma. J Neurosci Res. 1998;52(2):230–9. doi: 10.1002/(SICI)1097-4547(19980415)52:2<230::AID-JNR11>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
- 103.Lin W, et al. Endoplasmic reticulum stress modulates the response of myelinating oligodendrocytes to the immune cytokine interferon-gamma. J Cell Biol. 2005;169(4):603–12. doi: 10.1083/jcb.200502086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Vartanian T, et al. Interferon-gamma-induced oligodendrocyte cell death: implications for the pathogenesis of multiple sclerosis. Mol Med. 1995;1(7):732–43. [PMC free article] [PubMed] [Google Scholar]
- 105.Pouly S, et al. Interferon-gamma modulates human oligodendrocyte susceptibility to Fas-mediated apoptosis. J Neuropathol Exp Neurol. 2000;59(4):280–6. doi: 10.1093/jnen/59.4.280. [DOI] [PubMed] [Google Scholar]
- 106.Molina-Holgado E, et al. LPS/IFN-gamma cytotoxicity in oligodendroglial cells: role of nitric oxide and protection by the anti-inflammatory cytokine IL-10. Eur J Neurosci. 2001;13(3):493–502. doi: 10.1046/j.0953-816x.2000.01412.x. [DOI] [PubMed] [Google Scholar]
- 107.Buntinx M, et al. Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFN-gamma and TNF-alpha on apoptosis. J Neurosci Res. 2004;76(6):834–45. doi: 10.1002/jnr.20118. [DOI] [PubMed] [Google Scholar]
- 108.Mann SA, et al. Corticosteroids reverse cytokine-induced block of survival and differentiation of oligodendrocyte progenitor cells from rats. J Neuroinflammation. 2008;5:39. doi: 10.1186/1742-2094-5-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zhang X, et al. Cytokine toxicity to oligodendrocyte precursors is mediated by iron. Glia. 2005;52(3):199–208. doi: 10.1002/glia.20235. [DOI] [PubMed] [Google Scholar]
- 110.Merrill JE, et al. Microglial cell cytotoxicity of oligodendrocytes is mediated through nitric oxide. J Immunol. 1993;151(4):2132–41. [PubMed] [Google Scholar]
- 111.Merrill JE, Zimmerman RP. Natural and induced cytotoxicity of oligodendrocytes by microglia is inhibitable by TGF beta. Glia. 1991;4(3):327–31. doi: 10.1002/glia.440040311. [DOI] [PubMed] [Google Scholar]
- 112.Corbin JG, et al. Targeted CNS expression of interferon-gamma in transgenic mice leads to hypomyelination, reactive gliosis, and abnormal cerebellar development. Mol Cell Neurosci. 1996;7(5):354–70. doi: 10.1006/mcne.1996.0026. [DOI] [PubMed] [Google Scholar]
- 113.Balabanov R, et al. Suppressor of cytokine signaling 1 expression protects oligodendrocytes from the deleterious effects of interferon-gamma. J Neurosci. 2006;26(19):5143–52. doi: 10.1523/JNEUROSCI.0737-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.LaFerla FM, et al. Regional hypomyelination and dysplasia in transgenic mice with astrocyte-directed expression of interferon-gamma. J Mol Neurosci. 2000;15(1):45–59. doi: 10.1385/JMN:15:1:45. [DOI] [PubMed] [Google Scholar]
- 115.Lin W, et al. Interferon-gamma inhibits central nervous system remyelination through a process modulated by endoplasmic reticulum stress. Brain. 2006;129(Pt 5):1306–18. doi: 10.1093/brain/awl044. [DOI] [PubMed] [Google Scholar]
- 116.Panitch HS, et al. Treatment of multiple sclerosis with gamma interferon: exacerbations associated with activation of the immune system. Neurology. 1987;37(7):1097–102. doi: 10.1212/wnl.37.7.1097. [DOI] [PubMed] [Google Scholar]
- 117.Skurkovich S, et al. Randomized study of antibodies to IFN-gamma and TNF-alpha in secondary progressive multiple sclerosis. Mult Scler. 2001;7(5):277–84. doi: 10.1177/135245850100700502. [DOI] [PubMed] [Google Scholar]
- 118.Epstein LG, Prineas JW, Raine CS. Attachment of myelin to coated pits on macrophages in experimental allergic encephalomyelitis. J Neurol Sci. 1983;61(3):341–8. doi: 10.1016/0022-510x(83)90167-3. [DOI] [PubMed] [Google Scholar]
- 119.Field EJ, Raine CS. Experimental allergic encephalomyelitis. An electron microscopic study. Am J Pathol. 1966;49(3):537–53. [PMC free article] [PubMed] [Google Scholar]
- 120.Fleury HJ, et al. Further ultrastructural observations of virus morphogenesis and myelin pathology in JHM virus encephalomyelitis. Neuropathol Appl Neurobiol. 1980;6(3):165–79. doi: 10.1111/j.1365-2990.1980.tb00288.x. [DOI] [PubMed] [Google Scholar]
- 121.Tran EH, et al. Immune invasion of the central nervous system parenchyma and experimental allergic encephalomyelitis, but not leukocyte extravasation from blood, are prevented in macrophage-depleted mice. J Immunol. 1998;161(7):3767–75. [PubMed] [Google Scholar]
- 122.Bauer J, et al. The role of macrophages, perivascular cells, and microglial cells in the pathogenesis of experimental autoimmune encephalomyelitis. Glia. 1995;15(4):437–46. doi: 10.1002/glia.440150407. [DOI] [PubMed] [Google Scholar]
- 123.Hiremath MM, et al. Microglial/macrophage accumulation during cuprizone-induced demyelination in C57BL/6 mice. J Neuroimmunol. 1998;92(1–2):38–49. doi: 10.1016/s0165-5728(98)00168-4. [DOI] [PubMed] [Google Scholar]
- 124.Boyle EA, McGeer PL. Cellular immune response in multiple sclerosis plaques. Am J Pathol. 1990;137(3):575–84. [PMC free article] [PubMed] [Google Scholar]
- 125.Gonzalez JM, et al. Expression of a dominant negative IFN-gammareceptor on mouse oligodendrocytes. Glia. 2005;51(1):22–34. doi: 10.1002/glia.20182. [DOI] [PubMed] [Google Scholar]
- 126.Matute C, Perez-Cerda F. Multiple sclerosis: novel perspectives on newly forming lesions. Trends Neurosci. 2005;28(4):173–5. doi: 10.1016/j.tins.2005.01.006. [DOI] [PubMed] [Google Scholar]
- 127.Barnett MH, Prineas JW. Relapsing and remitting multiple sclerosis: pathology of the newly forming lesion. Ann Neurol. 2004;55(4):458–68. doi: 10.1002/ana.20016. [DOI] [PubMed] [Google Scholar]
- 128.Frohman EM, Racke MK, Raine CS. Multiple sclerosis--the plaque and its pathogenesis. N Engl J Med. 2006;354(9):942–55. doi: 10.1056/NEJMra052130. [DOI] [PubMed] [Google Scholar]
- 129.Liu Y, Cai Y, Zhang X. Induction of caspase-dependent apoptosis in cultured rat oligodendrocytes by murine coronavirus is mediated during cell entry and does not require virus replication. J Virol. 2003;77(22):11952–63. doi: 10.1128/JVI.77.22.11952-11963.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Liu Y, Pu Y, Zhang X. Role of the mitochondrial signaling pathway in murine coronavirus-induced oligodendrocyte apoptosis. J Virol. 2006;80(1):395–403. doi: 10.1128/JVI.80.1.395-403.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Liu Y, Zhang X. Expression of cellular oncogene Bcl-xL prevents coronavirus-induced cell death and converts acute infection to persistent infection in progenitor rat oligodendrocytes. J Virol. 2005;79(1):47–56. doi: 10.1128/JVI.79.1.47-56.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Liu Y, Zhang X. Murine coronavirus-induced oligodendrocyte apoptosis is mediated through the activation of the Fas signaling pathway. Virology. 2007;360(2):364–75. doi: 10.1016/j.virol.2006.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hauser KF, et al. HIV-1 Tat and morphine have interactive effects on oligodendrocyte survival and morphology. Glia. 2009;57(2):194–206. doi: 10.1002/glia.20746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Merabova N, et al. JC virus agnoprotein inhibits in vitro differentiation of oligodendrocytes and promotes apoptosis. J Virol. 2008;82(3):1558–69. doi: 10.1128/JVI.01680-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Albrecht PJ, et al. Astrocytes produce CNTF during the remyelination phase of viral-induced spinal cord demyelination to stimulate FGF-2 production. Neurobiol Dis. 2003;13(2):89–101. doi: 10.1016/s0969-9961(03)00019-6. [DOI] [PubMed] [Google Scholar]
- 136.Messersmith DJ, et al. Fibroblast growth factor 2 (FGF2) and FGF receptor expression in an experimental demyelinating disease with extensive remyelination. J Neurosci Res. 2000;62(2):241–56. doi: 10.1002/1097-4547(20001015)62:2<241::AID-JNR9>3.0.CO;2-D. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hatch MN, et al. Endogenous remyelination is induced by transplant rejection in a viral model of multiple sclerosis. J Neuroimmunol. 2009 doi: 10.1016/j.jneuroim.2009.05.002. [DOI] [PubMed] [Google Scholar]
- 138.Bruck W, Kuhlmann T, Stadelmann C. Remyelination in multiple sclerosis. J Neurol Sci. 2003;206(2):181–5. doi: 10.1016/s0022-510x(02)00191-0. [DOI] [PubMed] [Google Scholar]
- 139.Solanky M, et al. Proliferating oligodendrocytes are present in both active and chronic inactive multiple sclerosis plaques. J Neurosci Res. 2001;65(4):308–17. doi: 10.1002/jnr.1155. [DOI] [PubMed] [Google Scholar]
- 140.Barres BA, et al. Cell death in the oligodendrocyte lineage. J Neurobiol. 1992;23(9):1221–30. doi: 10.1002/neu.480230912. [DOI] [PubMed] [Google Scholar]
- 141.Barres BA, et al. Multiple extracellular signals are required for long-term oligodendrocyte survival. Development. 1993;118(1):283–95. doi: 10.1242/dev.118.1.283. [DOI] [PubMed] [Google Scholar]
- 142.Gard AL, et al. Astroglial control of oligodendrocyte survival mediated by PDGF and leukemia inhibitory factor-like protein. Development. 1995;121(7):2187–97. doi: 10.1242/dev.121.7.2187. [DOI] [PubMed] [Google Scholar]
- 143.Mayer M, Bhakoo K, Noble M. Ciliary neurotrophic factor and leukemia inhibitory factor promote the generation, maturation and survival of oligodendrocytes in vitro. Development. 1994;120(1):143–53. doi: 10.1242/dev.120.1.143. [DOI] [PubMed] [Google Scholar]
- 144.Zhang Y, et al. Interleukin-11 potentiates oligodendrocyte survival and maturation, and myelin formation. J Neurosci. 2006;26(47):12174–85. doi: 10.1523/JNEUROSCI.2289-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Li Y, et al. Coronavirus neurovirulence correlates with the ability of the virus to induce proinflammatory cytokine signals from astrocytes and microglia. J Virol. 2004;78(7):3398–406. doi: 10.1128/JVI.78.7.3398-3406.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Marriott MP, et al. Leukemia inhibitory factor signaling modulates both central nervous system demyelination and myelin repair. Glia. 2008;56(6):686–98. doi: 10.1002/glia.20646. [DOI] [PubMed] [Google Scholar]
- 147.Butzkueven H, et al. Endogenous leukemia inhibitory factor production limits autoimmune demyelination and oligodendrocyte loss. Glia. 2006;53(7):696–703. doi: 10.1002/glia.20321. [DOI] [PubMed] [Google Scholar]
- 148.Butzkueven H, et al. LIF receptor signaling limits immune-mediated demyelination by enhancing oligodendrocyte survival. Nat Med. 2002;8(6):613–9. doi: 10.1038/nm0602-613. [DOI] [PubMed] [Google Scholar]
- 149.van Oosten BW, et al. Increased MRI activity and immune activation in two multiple sclerosis patients treated with the monoclonal anti-tumor necrosis factor antibody cA2. Neurology. 1996;47(6):1531–4. doi: 10.1212/wnl.47.6.1531. [DOI] [PubMed] [Google Scholar]
- 150.Segal BM, et al. Repeated subcutaneous injections of IL12/23 p40 neutralising antibody, ustekinumab, in patients with relapsing-remitting multiple sclerosis: a phase II, double-blind, placebo-controlled, randomised, dose-ranging study. Lancet Neurol. 2008;7(9):796–804. doi: 10.1016/S1474-4422(08)70173-X. [DOI] [PubMed] [Google Scholar]

