Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters










Publication year range
1.
J Virol ; 89(18): 9299-312, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26136579

ABSTRACT

UNLABELLED: Myd88 signaling is critical to the control of numerous central nervous system (CNS) infections by promoting both innate and adaptive immune responses. Nevertheless, the extent to which Myd88 regulates type I interferon (IFN) versus proinflammatory factors and T cell function, as well as the anatomical site of action, varies extensively with the pathogen. CNS infection by neurotropic coronavirus with replication confined to the brain and spinal cord induces protective IFN-α/ß via Myd88-independent activation of melanoma differentiation-associated gene 5 (MDA5). However, a contribution of Myd88-dependent signals to CNS pathogenesis has not been assessed. Infected Myd88(-/-) mice failed to control virus, exhibited enhanced clinical disease coincident with increased demyelination, and succumbed to infection within 3 weeks. The induction of IFN-α/ß, as well as of proinflammatory cytokines and chemokines, was impaired early during infection. However, defects in both IFN-α/ß and select proinflammatory factors were rapidly overcome prior to T cell recruitment. Myd88 deficiency also specifically blunted myeloid and CD4 T cell recruitment into the CNS without affecting CD8 T cells. Moreover, CD4 T cells but not CD8 T cells were impaired in IFN-γ production. Ineffective virus control indeed correlated most prominently with reduced antiviral IFN-γ in the CNS of Myd88(-/-) mice. The results demonstrate a crucial role for Myd88 both in early induction of innate immune responses during coronavirus-induced encephalomyelitis and in specifically promoting protective CD4 T cell activation. In the absence of these responses, functional CD8 T cells are insufficient to control viral spread within the CNS, resulting in severe demyelination. IMPORTANCE: During central nervous system (CNS) infections, signaling through the adaptor protein Myd88 promotes both innate and adaptive immune responses. The extent to which Myd88 regulates antiviral type I IFN, proinflammatory factors, adaptive immunity, and pathology is pathogen dependent. These results reveal that Myd88 protects from lethal neurotropic coronavirus-induced encephalomyelitis by accelerating but not enhancing the induction of IFN-α/ß, as well as by promoting peripheral activation and CNS accumulation of virus-specific CD4 T cells secreting IFN-γ. By controlling both early innate immune responses and CD4 T cell-mediated antiviral IFN-γ, Myd88 signaling limits the initial viral dissemination and is vital for T cell-mediated control of viral loads. Uncontrolled viral replication in the absence of Myd88 leads to severe demyelination and pathology despite overall reduced inflammatory responses. These data support a vital role of Myd88 signaling in protective antimicrobial functions in the CNS by promoting proinflammatory mediators and T cell-mediated IFN-γ production.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Immunity, Cellular , Immunity, Innate , Maus Elberfeld virus/immunology , Myeloid Differentiation Factor 88/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Encephalitis, Viral/genetics , Encephalitis, Viral/pathology , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Maus Elberfeld virus/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics
2.
J Neuroimmunol ; 185(1-2): 57-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17343922

ABSTRACT

Although the central nervous system (CNS) is thought to be immunoprivileged, under special circumstances it can produce antibody. Antibody production within the CNS, called intrathecal antibody production (ITAbP), is a prominent feature of neurological infections and inflammatory diseases, and is thought to possibly contribute to disease in multiple sclerosis (MS), but it has not been extensively studied. We investigated ITAbP in a viral model of MS. ELISpot, real-time RT-PCR for IgG mRNA in CNS tissue, and CSF analysis were used to assess ITAbP in two types of SJL mice infected with one of two strains of Theiler's murine encephalomyelitis virus (TMEV). The amplitude of ITAbP increased during the first 4 months of infection. TMEV viral load remained high during the course of the infection, which likely was the main stimulus for ITAbP, since within samples of infected CNS tissues, levels of IgG gene expression were highly correlated with viral RNA levels, and a large percentage of intrathecally produced antibody was directed against TMEV. This study provides the first extensive analysis of ITAbP in TMEV infection, and demonstrates that, in this animal model of MS, antibody production within the CNS is likely driven by the presence of the causative pathogen.


Subject(s)
Antibodies, Viral/cerebrospinal fluid , Central Nervous System/immunology , Immunoglobulin G/cerebrospinal fluid , Maus Elberfeld virus/immunology , Multiple Sclerosis/immunology , Animals , Antibodies, Viral/biosynthesis , Antibody Formation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/biosynthesis , Mice , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
3.
J Virol ; 80(14): 7146-58, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809320

ABSTRACT

The innate and adaptive immune responses have evolved distinct strategies for controlling different viral pathogens. Encephalomyocarditis virus (EMCV) is a picornavirus that can cause paralysis, diabetes, and myocarditis within days of infection. The optimal innate immune response against EMCV in vivo requires CD1d. Interaction of antigen-presenting cell CD1d with distinct natural killer T-cell ("NKT") populations can induce rapid gamma interferon (IFN-gamma) production and NK-cell activation. The T-cell response of CD1d-deficient mice (lacking all NKT cells) against acute EMCV infection was further studied in vitro and in vivo. EMCV persisted at higher levels in CD1d-knockout (KO) splenocyte cultures infected in vitro. Furthermore, optimal resistance to repeat cycles of EMCV infection in vitro was also shown to depend on CD1d. However, this was not reflected in the relative levels of NK-cell activation but rather by the responses of both CD4(+) and CD8(+) T-cell populations. Repeated EMCV infection in vitro induced less IFN-gamma and alpha interferon (IFN-alpha) from CD1d-deficient splenocytes than with the wild type. Furthermore, the level of EMCV replication in wild-type splenocytes was markedly and specifically increased by addition of blocking anti-CD1d antibody. Depletion experiments demonstrated that dendritic cells contributed less than the combination of NK and NKT cells to anti-EMCV responses and that none of these cell types was the main source of IFN-alpha. Finally, EMCV infection in vivo produced higher levels of viremia in CD1d-KO mice than in wild-type animals, coupled with significantly less lymphocyte activation and IFN-alpha production. These results point to the existence of a previously unrecognized mechanism of rapid CD1d-dependent stimulation of the antiviral adaptive cellular immune response.


Subject(s)
Antigens, CD1/immunology , CD4-Positive T-Lymphocytes/immunology , Cardiovirus Infections/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Maus Elberfeld virus/immunology , Acute Disease , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigen Presentation/immunology , Antigens, CD1/genetics , Antigens, CD1d , CD4-Positive T-Lymphocytes/virology , Cardiovirus Infections/genetics , Diabetes Mellitus/immunology , Diabetes Mellitus/virology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-alpha/immunology , Interferon-gamma/immunology , Killer Cells, Natural/virology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Maus Elberfeld virus/genetics , Mice , Mice, Knockout , Myocarditis/immunology , Myocarditis/virology , Paralysis/immunology , Paralysis/virology , Viremia/genetics , Viremia/immunology , Virus Replication/drug effects , Virus Replication/genetics , Virus Replication/immunology
4.
J Virol ; 73(12): 9944-51, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10559307

ABSTRACT

Herein, we report that Autographa californica nucleopolyhedrovirus, a member of the Baculoviridae family, is capable of stimulating antiviral activity in mammalian cells. Baculoviruses are not pathogenic to mammalian cells. Nevertheless, live baculovirus is shown here to induce interferons (IFN) from murine and human cell lines and induces in vivo protection of mice from encephalomyocarditis virus infection. Monoclonal antibodies specific for the baculovirus envelope gp67 neutralize baculovirus-dependent IFN production. Moreover, UV treatment of baculovirus eliminates both infectivity and IFN-inducing activity. In contrast, the IFN-inducing activity of the baculovirus was unaffected by DNase or RNase treatment. These data demonstrate that IFN production can be induced in mammalian cells by baculovirus even though the cells fail to serve as a natural host for an active viral infection. Baculoviruses, therefore, provide a novel model in which to study at least one alternative mechanism for IFN induction in mammalian cells.


Subject(s)
Interferon-alpha/immunology , Interferon-beta/immunology , Nucleopolyhedroviruses/immunology , Animals , Antibodies, Monoclonal/immunology , COS Cells , Cardiovirus Infections/immunology , Cardiovirus Infections/prevention & control , DNA/immunology , Female , Humans , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Lipopolysaccharides/immunology , Mammals , Maus Elberfeld virus/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Moths/virology , Neutralization Tests , RNA, Double-Stranded/immunology
5.
Eur J Immunol ; 25(6): 1589-93, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7614986

ABSTRACT

Theiler's virus causes, in the susceptible SJL/J mouse, a chronic demyelinating disease that resembles multiple sclerosis. Demyelination is at least in part immune mediated and coincides with the infiltration of the central nervous system by T lymphocytes. We analyzed the repertoire of the T cell receptor (TcR) beta-chain for each V beta-J beta combination, in spinal cord and spleen T cells of infected animals. All V beta families were detected in spinal cord as well as in spleen, as if the central nervous system were undiscriminantly invaded by T cells in the infected animals. Some T cells, defined by specific V beta-J beta combinations, were expanded in the spinal cord but not in the spleen, most probably because of an antigen-driven response. The TcR beta-chain repertoire of infiltrating T cells was the same at the onset of demyelination as when the disease was full-blown. These results provide the first description of the repertoire of the T cells which infiltrate the central nervous system during the course of this disease.


Subject(s)
Maus Elberfeld virus/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Cell Movement , Female , Mice , Receptors, Antigen, T-Cell, alpha-beta/analysis , Spinal Cord/immunology , Spleen/immunology
6.
Microb Pathog ; 18(1): 11-27, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7783595

ABSTRACT

Intracerebral infection with the DA strain of Theiler's murine encephalomyelitis virus induces a chronic demyelinating disease in SJL/J mice. Intraperitoneal inoculation with either the wild-type DA virus or an attenuated variant virus of DA, H7A6-2, results in protection from development of chronic demyelinating disease. Protective anti-viral immune responses result in reduced viral titers and decreased inflammation in the central nervous system within the first week following intracerebral challenge with virus. Development of protective immunity requires the presence of B cells and CD4+ T cells but does not require CD8+ T cells. High titers of serum anti-viral IgG and neutralizing antibodies are induced following the intraperitoneal inoculation with the DA virus or H7A6-2 virus prior to challenge. While protection could not be transferred with immune serum from DA virus-infected mice or neutralizing monoclonal antibodies, protection was correlated with increased numbers of DA virus-specific plasma cells in the central nervous system within the first week following intracerebral challenge. Protected mice also had enhanced levels of anti-DA virus IgG and neutralizing antibodies in the cerebral spinal fluid by 1 week following intracerebral challenge with DA virus. Thus, we conclude that vaccination with live virus results in protection from chronic demyelinating disease by inducing immune responses which are manifested in the central nervous system and rapidly clear infection after intracerebral challenge with DA virus.


Subject(s)
Cardiovirus Infections/prevention & control , Central Nervous System/immunology , Demyelinating Diseases/prevention & control , Maus Elberfeld virus/immunology , Viral Vaccines/therapeutic use , Animals , B-Lymphocytes/immunology , Brain/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/pathology , Central Nervous System/virology , Female , Immunity, Cellular/immunology , Lymphocyte Depletion , Mice , Mice, Inbred Strains , Neutralization Tests , Plasma Cells , Spinal Cord/immunology , Vaccines, Attenuated/therapeutic use
7.
Int Immunol ; 6(5): 721-30, 1994 May.
Article in English | MEDLINE | ID: mdl-8080842

ABSTRACT

Efficient immunologic tolerance, defined as antigen-specific unresponsiveness, can be peripherally induced by the i.v. injection of syngeneic splenocytes coupled with antigen using ethylene carbodiimide (ECDI). We have previously reported that unresponsiveness induced via i.v. injection of syngeneic splenocytes coupled with intact, UV-inactivated Theiler's murine encephalomyelitis virus (TMEV-SP) resulted in 'split tolerance'. Both virus-specific delayed-type hypersensitivity and IgG2a levels were inhibited, whereas IgG1 levels were increased when compared with sham tolerized controls. In the present report we demonstrate that tolerance induced by i.v. injection of TMEV-coupled splenocytes resulted in antigen-specific inhibition of T cell proliferation, as well as IL-2 and IFN-gamma production in response to both whole TMEV and the immunodominant viral epitope. Additionally, tolerance induction resulted in abrogation of Th1-derived [IL-2, IFN-gamma and LT/tumor necrosis factor-beta (TNF-beta)] cytokine mRNA expression in response to in vitro stimulation with UV-inactivated TMEV as determined by reverse transcriptase polymerase chain reaction. In contrast, expression of Th2-derived (IL-4, IL-6 and IL-10) cytokine mRNA was not affected in tolerized mice. Tolerance functioned directly at the level of CD4+ Th1 cells at both the induction and effector limbs as depletion of CD8+ T cells both prior to in vivo tolerization or in vitro culture had no effect on inhibition of Th1-specific responses. The mechanism of in vivo tolerance induction appeared to be anergy of CD4+ Th1 cells since IL-2, IFN-gamma and LT/TNF-beta mRNA expression as well as virus-specific proliferative responses could be restored by addition of rIL-2 to in vitro cultures of tolerant, CD4+ Th1 populations. These results suggest that in vivo 'split tolerance' induced by i.v. injection of ECDI-fixed, antigen-coupled splenocytes involves anergy of TMEV-specific, CD4+ Th1 lymphocytes and concomitant priming of Th2 cells. The induction of antigen-specific, in vivo anergy has important implications in the design of therapeutic strategies for immunopathologic diseases mediated by Th1 lymphocytes, especially T cell-mediated autoimmune disorders.


Subject(s)
Clonal Anergy , Cytokines/biosynthesis , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Antigens, Viral/immunology , CD4 Antigens , Down-Regulation , Female , Immunodominant Epitopes/immunology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Maus Elberfeld virus/immunology , Mice , Molecular Sequence Data , RNA, Messenger/metabolism , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
8.
Eur J Immunol ; 23(9): 2287-93, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8370406

ABSTRACT

Intracerebral inoculation of Theiler's murine encephalomyelitis virus (TMEV) results in immune-mediated demyelination in susceptible mouse strains. The histology of TMEV-induced demyelination is similar to that seen in patients suffering from multiple sclerosis. It was previously shown that the susceptibility of mice to TMEV-induced demyelination in certain strain combinations is closely associated with the major histocompatibility complex (MHC) class I locus. Here we examine disease susceptibility of beta 2-microglobulin (beta 2M)-deficient transgenic mice lacking class I expression and functional CD8+ T cells. In contrast to TMEV-infected parental C57BL/6 mice, the transgenics develop high levels of virus-specific DTH and T cell proliferation accompanied by an increased frequency of central nervous system (CNS) demyelinating lesions. However, clinical signs of demyelination were not noted. Neither antibody titer nor viral persistence were significantly affected in the beta 2M-deficient mice. These results suggest that in the absence of functional class I/CD8+ cells, the class II-restricted T cell response to TMEV is enhanced and CNS pathogenesis is heightened, although the level is not severe enough to result in clinical disease. When the TMEV-infected mice were subcutaneously immunized with virus, however, the beta 2M-deficient mice displayed clinical symptoms. Therefore, our results strongly suggest that CD8+ T cells do not directly contribute to CNS demyelination. In contrast, such T cells appear to be primarily involved in down-regulation of a potentially damaging CD4+ T cell response in resistant animals, although some of the T cells may play a role in clearing viral persistence in the CNS, resulting in the protection of the host from viral demyelination.


Subject(s)
Antigens, Viral/immunology , Demyelinating Diseases/immunology , Histocompatibility Antigens Class I/physiology , Maus Elberfeld virus/immunology , T-Lymphocytes/immunology , beta 2-Microglobulin/deficiency , Animals , Antibodies, Viral/biosynthesis , Brain/microbiology , Cells, Cultured , Disease Susceptibility , Enterovirus Infections/immunology , Hypersensitivity, Delayed , Immunization , Lymphocyte Activation , Maus Elberfeld virus/isolation & purification , Mice , Mice, Inbred C57BL
9.
J Immunol ; 151(1): 266-76, 1993 Jul 01.
Article in English | MEDLINE | ID: mdl-8326128

ABSTRACT

Intracerebral infection of susceptible strains of mice with Theiler's virus, a picornavirus, results in central nervous system demyelination, which is similar to multiple sclerosis. Immunogenetic experiments indicate that the MHC (H-2) and, in particular, the D region that controls class I-restricted immune responses, is an important determinant to development of demyelination. We tested whether disruption of beta 2-microglobulin (beta 2-m) would abrogate resistance to demyelinating disease normally observed in H-2b mice. All (C57BI/6 x 129)F3 mice transgenic for homozygous beta 2-m gene disruption (-/-) developed chronic demyelination after Theiler's murine encephalomyelitis virus infection, whereas none of the infected littermates with normal expression of class I MHC (beta 2-m, +/+) developed demyelination. Demyelinated lesions showed class II MHC expression, macrophages, and TNF but no class I MHC expression or CD8+ T cells. No correlation was observed between development of demyelination and delayed-type hypersensitivity responses to virus Ag. Despite the presence of demyelinating lesions, none of the infected beta 2-m (-/-) mice developed neurologic deficits. Infectious virus and virus Ag persisted in the central nervous systems of infected beta 2-m (-/-) mice but not in beta 2-m (+/+) mice. These experiments support the hypothesis that a class I immune response mediated by CD8+ T cells is important in resistance to Theiler's murine encephalomyelitis virus-induced demyelination. Development of chronic neurologic deficits as observed in immunocompetent susceptible strains of mice may be dependent on the presence of class I MHC and CD8+ T cells.


Subject(s)
Demyelinating Diseases/immunology , Enterovirus Infections/immunology , Maus Elberfeld virus/immunology , T-Lymphocyte Subsets/immunology , beta 2-Microglobulin/deficiency , Animals , Antibodies, Viral/biosynthesis , Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , Demyelinating Diseases/pathology , Gene Expression , Genes, MHC Class I , Genes, MHC Class II , Hypersensitivity, Delayed/immunology , Mice , Mice, Transgenic , Mutagenesis, Insertional , Skin/immunology
10.
Cell Immunol ; 149(2): 237-47, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8330309

ABSTRACT

Increasing evidence shows that interleukin-1 (IL-1) contributes to inflammatory processes, such as experimental allergic encephalomyelitis (EAE) and virus-induced demyelination, inside the central nervous system (CNS). Using primary cultures of mouse astrocytes, we show that these glial cells can be induced to produce IL-1 alpha when infected with Theiler's murine encephalomyelitis virus (TMEV). This was true for astrocytes from SJL/J mice, a strain susceptible to TMEV-induced demyelination. Conversely, BALB/c astrocytes, derived from animals genetically resistant to demyelination, did not produce IL-1 alpha in detectable amounts. Therefore, a differential IL-1 gene expression, which is strain specific, is demonstrated after TMEV infection in astrocytes. The release of IL-1 alpha by SJL astrocytes was studied from kinetic, infectivity, and immunochemical points of view. Since IL-1 plays a critical role in the immune response, its production by astrocytes in some strains of mice may contribute to virus-induced susceptibility and to inflammation associated with this experimental model of multiple sclerosis.


Subject(s)
Astrocytes/immunology , Interleukin-1/immunology , Maus Elberfeld virus/immunology , Multiple Sclerosis/immunology , Animals , Astrocytes/metabolism , Base Sequence , Cells, Cultured , Disease Models, Animal , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Inflammation/immunology , Interleukin-1/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Sequence Data
11.
Am J Pathol ; 143(1): 221-9, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8317548

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) infection of mice can produce a biphasic disease of the central nervous system (CNS). Most susceptible strains of mice survive the acute infection and develop a chronic demyelinating disease. In this report, we analyzed the routes of spread of TMEV within the CNS of nude mice and target sites eventually infected in the CNS. Compared to the immunocompetent mouse, in which an antiviral immune response is mounted but virus persists, the nude mouse develops a severe encephalomyelitis due to the lack of functional T lymphocytes and provides a useful model for the study of viral dissemination. We demonstrated, by immunohistochemistry, the presence of viral antigen in defined regions of the CNS, corresponding to various structures of the limbic system. In addition, we found a different time course for viral spread using two different sites of intracerebral inoculation, ie, via the olfactory bulb or the cortex. Limbic structures were rapidly infected following olfactory bulb infection and then showed a decrease in viral load, presumably due to loss of target neurons. Using either route of infection, the virus was able to disseminate to similar regions. These results indicate that limbic structures and their connections are very important for the spread of TMEV in the brain. In the spinal cord, not only neuronal but hematogenous pathways were suspected to be involved in the dissemination of Theiler's virus.


Subject(s)
Brain Diseases/microbiology , Enterovirus Infections/transmission , Limbic System/microbiology , Maus Elberfeld virus , Olfactory Pathways/microbiology , Animals , Antigens, Viral/analysis , Demyelinating Diseases/etiology , Female , Hippocampus/immunology , Hippocampus/microbiology , Maus Elberfeld virus/immunology , Mice , Mice, Nude , Neuroglia/cytology , Neuroglia/immunology , Spinal Cord/immunology , Spinal Cord/microbiology , Substantia Nigra/immunology , Substantia Nigra/microbiology , Time Factors
12.
J Neuroimmunol ; 45(1-2): 67-73, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8331166

ABSTRACT

Intracerebral (i.c.) inoculation of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) results in a demyelinating disease similar to human multiple sclerosis (MS). Mice develop a strong immune response to TMEV and the disease is believed to be immune-mediated. In order to investigate the effects of the immune response to TMEV on the course of demyelination, we immunized host mice with UV-inactivated TMEV at various time periods in relation to intracerebral inoculation with live TMEV. Here, we show that subcutaneous immunization of mice with TMEV prior to infection with virus is able to protect susceptible, SJL/J mice from demyelinating disease. This protective effect appears to be long-lasting; immunization greater than 90 days prior to i.c. inoculation of the virus protects mice from subsequent infection. However, immunization of mice after i.c. infection with TMEV does not confer protection, but rather exacerbates the disease symptoms. Thus, this system offers a model for studying viral capsid proteins and/or epitopes which are involved in either protection from disease or immune-mediated pathogenesis leading to myelin destruction in susceptible mice.


Subject(s)
Demyelinating Diseases/prevention & control , Immunization , Maus Elberfeld virus/immunology , Animals , Demyelinating Diseases/physiopathology , Disease Susceptibility , Maus Elberfeld virus/radiation effects , Mice , Mice, Inbred Strains , Myelin Sheath/physiology , Ovalbumin/immunology , Time Factors , Ultraviolet Rays , Vaccines, Inactivated
13.
Eur J Immunol ; 23(1): 46-55, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8419186

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) produces a chronic, inflammatory demyelinating disease in susceptible mouse strains that is used as a model for multiple sclerosis. Because disease susceptibility correlates temporally with the development of virus-specific delayed-type hypersensitivity (DTH) responses, we studied methods and mechanisms by which virus-specific DTH could be specifically inhibited. The intravenous injection of UV-inactivated TMEV coupled to syngeneic splenocytes via a carbodiimide linkage (TMEV-SP), prior to immunization, induced a significant degree of tolerance in virus-specific helper (Th) cells as determined by decreased DTH and T cell proliferative responses, and decreased interleukin (IL)-2 and interferon (IFN)-gamma protein and mRNA levels. In contrast to the reduced levels of Th1-specific lymphokine mRNA levels, IL-4-specific mRNA levels in response to virus stimulation were not affected in tolerant mice. Surprisingly, the total anti-TMEV antibody response in DTH tolerant mice was enhanced 20-100-fold over sham-tolerized controls and was composed of reduced levels of anti-virus IgG2a, but dramatically increased levels of anti-virus IgG1. The "split-tolerance" was antigen specific, dependent on the concentrations of TMEV and carbodiimide used in the coupling procedure, and varied with the number of coupled syngeneic splenocytes administered. The fixative effects of carbodiimide on antigen-presenting function were necessary for the induction of DTH tolerance with TMEV-SP, since intravenous administration of virus coupled to splenocytes via a biotin-avidin linkage led to enhanced virus-specific antibody responses, but was unable to inhibit DTH unless concomitantly fixed with carbodiimide. Collectively, the data indicate that Th1 cells (mediating DTH, IL-2 and IFN-gamma production, and helper function for IgG2a production) were specifically anergized, with concomitant stimulation of Th2 cells (producing IL-4 and mediating helper function for IgG1 antibody production).


Subject(s)
Immune Tolerance , Maus Elberfeld virus/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Viral/analysis , Female , Hypersensitivity, Delayed/immunology , Immunoglobulin G/analysis , Lymphokines/genetics , Mice , RNA, Messenger/analysis
14.
J Virol ; 67(1): 589-92, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8416386

ABSTRACT

Theiler's virus, a murine picornavirus, persists in the central nervous systems of susceptible mice and induces a chronic demyelinating disease. Susceptibility or resistance to this disease is controlled in part by the H2-D locus of the major histocompatibility complex (MHC). For this reason, it has been proposed that CD8+ class I-restricted cytotoxic T cells play a main role in the pathogenesis of this viral infection. We recently reported the existence of anti-virus CD8+ cytotoxic T cells in the course of Theiler's virus infection. In the present study, we examined the role of these effector cells in mice in which the beta 2-microglobulin gene had been disrupted. These mice fail to express class I MHC molecules and therefore lack CD8+ T cells. The mice are derived from a C57BL/6 x 129/Ola cross and are H-2b, a haplotype associated with resistance to Theiler's virus infection. beta 2-Microglobulin-deficient mice (beta 2m-/-mice) failed to clear the virus, developed demyelination, and, interestingly, did not succumb to early infection. These results demonstrate that CD8+ T cells are required to clear Theiler's virus infection. In contrast with a current hypothesis, they also demonstrate that CD8+ T cells are not major mediators of the demyelinating disease.


Subject(s)
CD8 Antigens/immunology , Enterovirus Infections/immunology , Maus Elberfeld virus/immunology , Mice, Mutant Strains/immunology , beta 2-Microglobulin/deficiency , Animals , Crosses, Genetic , Genes, MHC Class I , Haplotypes , Major Histocompatibility Complex/immunology , Maus Elberfeld virus/pathogenicity , Mice , Mice, Inbred C57BL , Spinal Cord/pathology , Spinal Cord/ultrastructure , T-Lymphocytes, Cytotoxic/immunology , Virulence , beta 2-Microglobulin/immunology
15.
Rev Sci Tech ; 11(4): 1071-7, 1992 Dec.
Article in French | MEDLINE | ID: mdl-1339064

ABSTRACT

The results of a serological survey of a free-living population of meadow voles (Microtus pennsylvanicus) in Pinawa, Manitoba (Canada) showed that these animals possessed antibodies to six of the eleven viruses tested for, namely: reovirus type 3, murine encephalomyelitis agent, ectromelia virus, murine adenovirus, murine hepatitis virus and lymphocytic choriomeningitis virus. The significant increase in the number of individuals possessing specific antibodies suggests that these viruses, or related viruses, may be responsible for the decline in the population studied.


Subject(s)
Antibodies, Viral/blood , Arvicolinae , Rodent Diseases/epidemiology , Virus Diseases/veterinary , Adenoviridae/immunology , Adenoviridae Infections/epidemiology , Adenoviridae Infections/immunology , Adenoviridae Infections/veterinary , Animals , Ectromelia virus/immunology , Ectromelia, Infectious/epidemiology , Ectromelia, Infectious/immunology , Enterovirus Infections/epidemiology , Enterovirus Infections/immunology , Enterovirus Infections/veterinary , Hepatitis, Viral, Animal/epidemiology , Hepatitis, Viral, Animal/immunology , Incidence , Lymphocytic Choriomeningitis/epidemiology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mammalian orthoreovirus 3/immunology , Manitoba/epidemiology , Maus Elberfeld virus/immunology , Murine hepatitis virus/immunology , Reoviridae Infections/epidemiology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Rodent Diseases/immunology , Virus Diseases/epidemiology , Virus Diseases/immunology
16.
Mayo Clin Proc ; 67(9): 829-38, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1434926

ABSTRACT

Chronic infection of susceptible strains of mice with Theiler's murine encephalomyelitis virus (TMEV) results in central nervous system (CNS) demyelination similar to multiple sclerosis. Demyelination induced by TMEV is mediated, in part, by class I-restricted CD8+ T lymphocytes. For these T cells to function, they must recognize virus-infected CNS targets in the presence of class I major histocompatibility complex (MHC) antigen. Therefore, we studied in vivo expression of class I MHC antigen and viral antigen-RNA in prototypic mouse strains that are susceptible (SJL/J) or resistant (C57BL/10SNJ) to TMEV-induced demyelination. In brains of resistant mice, viral antigen-RNA expression peaked on day 3 after infection and was effectively diminished by day 5 such that few virus-infected cells were ever detected in the spinal cord. In contrast, susceptible mice demonstrated delay in clearance of TMEV from the brain and a subsequent increase and persistence of viral antigen-RNA in the spinal cord for as long as 277 days. Viral infection resulted in "upregulation" of class I MHC expression in the CNS. Class I MHC antigens were expressed as early as 1 day after infection in the choroid plexus of both strains of mice before detection of viral antigen or inflammation. In resistant mice, class I MHC expression predominated in the gray matter of the brain and spinal cord on day 7 after infection but returned to undetectable levels by day 28. In susceptible mice, class I MHC expression in the CNS persisted and was intense in the white matter of the spinal cord throughout chronic infection and demyelination. No class I MHC expression was detected in the CNS of uninfected mice. Coexpression of viral RNA and class I MHC antigen was demonstrated in CNS cells by using simultaneous in situ hybridization and immunoperoxidase technique. These results support the hypothesis that a class I-restricted immune response directed against virus-infected cells may be important in the mechanism of demyelination.


Subject(s)
Central Nervous System/microbiology , Disease Models, Animal , Histocompatibility Antigens Class I/analysis , Maus Elberfeld virus/immunology , Multiple Sclerosis/pathology , RNA, Viral/analysis , Animals , Antigens, Viral/analysis , Central Nervous System/immunology , Central Nervous System/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/microbiology , Demyelinating Diseases/pathology , Disease Susceptibility , Enterovirus Infections/immunology , Enterovirus Infections/pathology , Female , Maus Elberfeld virus/genetics , Mice , Mice, Inbred Strains , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology
17.
Immunology ; 76(1): 133-9, 1992 May.
Article in English | MEDLINE | ID: mdl-1628891

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) is a picornavirus which induces a chronic demyelinating disease of the central nervous system (CNS) in certain susceptible mouse strains. Demyelination has been shown to result from immunopathological responses mediated by CD4+, major histocompatibility complex (MHC) class II-restricted T cells. As little or no class II is expressed in the normal mouse CNS, the ability of astrocytes to express these proteins and present antigen to T cells from TMEV-infected mice was investigated here. It is shown that astrocytes are capable of presenting TMEV to virus-specific T cells in vitro, and that this ability is dependent on prior induction of MHC class II by interferon-gamma (IFN-gamma) treatment. Unlike other viruses such as murine hepatitis virus-JHM (a coronavirus) and measles, TMEV is not capable of inducing class II on astrocytes directly. There is a correlation between the ease of class II induction on astrocytes from different mouse strains by IFN-gamma and mouse strain susceptibility to TMEV-induced demyelinating disease. These results suggest that following viral infection and initial T-cell infiltration into the CNS, class II induction on astrocytes is a key step allowing local antigen presentation and amplification of immunopathological responses within the CNS and hence the development of demyelinating disease.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Viral/immunology , Astrocytes/immunology , Enterovirus Infections/immunology , Maus Elberfeld virus/immunology , Animals , Cells, Cultured , Disease Susceptibility , Histocompatibility Antigens Class II/analysis , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred Strains , Recombinant Proteins , T-Lymphocytes/immunology
18.
Immunology ; 75(4): 652-8, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1350571

ABSTRACT

Inbred mouse strains differ in susceptibility to Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease. A strong correlation between disease susceptibility and delayed-type hypersensitivity (DTH) has been previously demonstrated, but no strong correlation between disease susceptibility and total anti-TMEV ELISA titres was shown. Since both DTH and IgG2a antibody production are regulated by CD4+ Th1 cells, we investigated three strains of mice to determine whether antivirus IgG2a antibody levels, like DTH in previous studies, correlated with disease susceptibility. Susceptible SJL/J, intermediately susceptible C3H/HeJ, and resistant C57BL/6 mice were infected intracerebrally (i.c.) with the BeAn strain of TMEV and monitored for clinical signs of demyelination and for levels of TMEV-specific antibody of different IgG subclasses using a particle concentration fluorescence immunoassay (PCFIA). Resistant C57BL/6 mice were found to have significantly lower concentrations of total anti-TMEV antibody than susceptible SJL/J mice and intermediately susceptible C3H/HeJ mice show variable antibody responses. A predominance of anti-TMEV IgG2a (Th1 regulated) antibody was seen in susceptible and intermediately susceptible mice, whereas resistant mice displayed a predominant anti-TMEV IgG1 (Th2 regulated) response accompanied by a marked deficiency of IgG2a. In contrast, immunization of C57BL/6 mice with UV-inactivated TMEV in adjuvant revealed that this strain was not defective either in its ability to generate high levels of anti-TMEV antibody or in its ability to produce IgG2a antibody. These results suggest that the antivirus IgG subclass profile is dependent upon the immunization route, virus viability and/or the use of adjuvant and that the levels of antivirus subclasses may be predictive of disease susceptibility.


Subject(s)
Antibodies, Viral/biosynthesis , CD4-Positive T-Lymphocytes/immunology , Enterovirus Infections/immunology , Immunoglobulin G/biosynthesis , Maus Elberfeld virus/immunology , Animals , Antibody Specificity/immunology , Disease Susceptibility , Female , Immunization , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains
19.
J Immunol ; 148(3): 921-7, 1992 Feb 01.
Article in English | MEDLINE | ID: mdl-1309845

ABSTRACT

Intracerebral infection of certain strains of mice with Theiler's virus results in chronic immune-mediated demyelination in spinal cord. We used mouse mutants with deletion of the V beta class of TCR genes to examine the role of TCR genes in this demyelinating disease which is similar to multiple sclerosis. Quantitative analysis of spinal cord lesions demonstrated a markedly increased number and extent of demyelinated lesions in persistently infected RIII S/J mice which have a massive deletion of the TCR V beta-chain (V beta 5.2, V beta 8.3, V beta 5.1, V beta 8.2, V beta 5.3, V beta 8.1, V beta 13, V beta 12, V beta 11, V beta 9, V beta 6, V beta 15, V beta 17) compared with B10.RIII mice which are of identical MHC haplotype (H-2r) but have normal complement of V beta TCR genes. In contrast, infection of C57L (H-2b) or C57BR (H-2k) mice which have deletion of the V beta TCR genes (V beta 5.2, V beta 8.3, V beta 5.1, V beta 8.2, V beta 5.3, V beta 8.1, V beta 13, V beta 12, V beta 11, and V beta 9) resulted in few demyelinating lesions. Genetic segregation analysis of (B10.RIII x RIII S/J) x RIII S/J backcrossed mice and (B10.RIII x RIII S/J) F2 mice demonstrated correlation of increased susceptibility to demyelination with deletion of TCR V beta genes. The increase in number of demyelinating lesions correlated with increase in number of virus-Ag+ cells in spinal cord. These experiments provide strong evidence that the structural diversity at the TCR beta-complex can influence susceptibility to virus-induced demyelination.


Subject(s)
Demyelinating Diseases/immunology , Enterovirus Infections/immunology , Maus Elberfeld virus/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Antigens, Viral/analysis , Chromosome Deletion , Demyelinating Diseases/microbiology , Demyelinating Diseases/pathology , Enterovirus Infections/pathology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Mice , Mice, Inbred Strains
20.
Neurology ; 42(2): 348-57, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1736164

ABSTRACT

Immunosuppression using cyclophosphamide or anti-T cell monoclonal antibodies (mAbs) directed at CD4 or CD8 promoted remyelination of CNS axons in the spinal cords of mice infected chronically with Theiler's virus. Treatment with a mAb directed at class II major histocompatibility gene products did not increase the extent of CNS remyelination. Following immunosuppressive treatment, quantitative morphometry revealed a five- to sevenfold increase in new myelin synthesis. Proliferating nervous system cells were identified at the edges of remyelinated lesions by their incorporation of [3H]thymidine. CNS remyelination occurred in mice depleted of selected subsets of T lymphocytes despite the local persistence of viral antigen. These findings indicate that CNS remyelination occurs as a normal consequence of primary myelin injury, but factors associated with immune T cells somehow impair remyelination. Interference with the function of immune T cells enhances CNS remyelination by oligodendrocytes. Similar depletion of immune T cells may allow for enhanced remyelination in the CNS of patients with chronic multiple sclerosis.


Subject(s)
Central Nervous System/immunology , Demyelinating Diseases/immunology , Immunosuppression Therapy , Myelin Sheath/physiology , Animals , Antigens, Viral/immunology , Autoradiography , Axons/immunology , Axons/physiology , Axons/ultrastructure , Cell Division/immunology , Central Nervous System/physiopathology , Chronic Disease , Demyelinating Diseases/microbiology , Demyelinating Diseases/physiopathology , Enterovirus Infections/immunology , Enterovirus Infections/physiopathology , Maus Elberfeld virus/immunology , Mice , Microscopy, Electron , Myelin Sheath/immunology , Myelin Sheath/ultrastructure , Neuroglia/immunology , Neuroglia/physiology , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL