1
|
Excessive Innate Immunity Steers Pathogenic Adaptive Immunity in the Development of Theiler's Virus-Induced Demyelinating Disease. Int J Mol Sci 2021; 22:ijms22105254. [PMID: 34067536 PMCID: PMC8156427 DOI: 10.3390/ijms22105254] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/06/2021] [Accepted: 05/13/2021] [Indexed: 01/05/2023] Open
Abstract
Several virus-induced models were used to study the underlying mechanisms of multiple sclerosis (MS). The infection of susceptible mice with Theiler’s murine encephalomyelitis virus (TMEV) establishes persistent viral infections and induces chronic inflammatory demyelinating disease. In this review, the innate and adaptive immune responses to TMEV are discussed to better understand the pathogenic mechanisms of viral infections. Professional (dendritic cells (DCs), macrophages, and B cells) and non-professional (microglia, astrocytes, and oligodendrocytes) antigen-presenting cells (APCs) are the major cell populations permissive to viral infection and involved in cytokine production. The levels of viral loads and cytokine production in the APCs correspond to the degrees of susceptibility of the mice to the TMEV-induced demyelinating diseases. TMEV infection leads to the activation of cytokine production via TLRs and MDA-5 coupled with NF-κB activation, which is required for TMEV replication. These activation signals further amplify the cytokine production and viral loads, promote the differentiation of pathogenic Th17 responses, and prevent cellular apoptosis, enabling viral persistence. Among the many chemokines and cytokines induced after viral infection, IFN α/β plays an essential role in the downstream expression of costimulatory molecules in APCs. The excessive levels of cytokine production after viral infection facilitate the pathogenesis of TMEV-induced demyelinating disease. In particular, IL-6 and IL-1β play critical roles in the development of pathogenic Th17 responses to viral antigens and autoantigens. These cytokines, together with TLR2, may preferentially generate deficient FoxP3+CD25- regulatory cells converting to Th17. These cytokines also inhibit the apoptosis of TMEV-infected cells and cytolytic function of CD8+ T lymphocytes (CTLs) and prolong the survival of B cells reactive to viral and self-antigens, which preferentially stimulate Th17 responses.
Collapse
|
2
|
Rapid Expansion of Virus-Specific CD4 + T Cell Types in the CNS of Susceptible Mice Infected with Theiler's Virus. Int J Mol Sci 2020; 21:ijms21207719. [PMID: 33086489 PMCID: PMC7588906 DOI: 10.3390/ijms21207719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 01/19/2023] Open
Abstract
The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) induces a T cell-mediated demyelinating disease. This system has been studied as a relevant infection model for multiple sclerosis (MS). Therefore, defining the type of T cell responses and their functions is critically important for understanding the relevant pathogenic mechanisms. In this study, we adoptively transferred naive VP2-specific TCR-Tg CD4+ T cells into syngeneic susceptible SJL mice and monitored the development of the disease and the activation and proliferation of CD4+ T cells during the early stages of viral infection. The preexisting VP2-specific naive CD4+ T cells promoted the pathogenesis of the disease in a dose-dependent manner. The transferred VP2-specific CD4+ T cells proliferated rapidly in the CNS starting at 2-3 dpi. High levels of FoxP3+CD4+ T cells were found in the CNS early in viral infection (3 dpi) and persisted throughout the infection. Activated VP2-specific FoxP3+CD4+ T cells inhibited the production of IFN-γ, but not IL-17, via the same VP2-specific CD4+ T cells without interfering in proliferation. Thus, the early presence of regulatory T cells in the CNS with viral infection may favor the induction of pathogenic Th17 cells over protective Th1 cells in susceptible mice, thereby establishing the pathogenesis of virus-induced demyelinating disease.
Collapse
|
3
|
Tritz ZP, Orozco RC, Malo CS, Ayasoufi K, Fain CE, Khadka RH, Goddery EN, Yokanovich LT, Settell ML, Hansen MJ, Jin F, Pavelko KD, Pease LR, Johnson AJ. Conditional Silencing of H-2D b Class I Molecule Expression Modulates the Protective and Pathogenic Kinetics of Virus-Antigen-Specific CD8 T Cell Responses during Theiler's Virus Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:1228-1238. [PMID: 32737149 DOI: 10.4049/jimmunol.2000340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection of the CNS is cleared in C57BL/6 mice by a CD8 T cell response restricted by the MHC class I molecule H-2Db The identity and function of the APC(s) involved in the priming of this T cell response is (are) poorly defined. To address this gap in knowledge, we developed an H-2Db LoxP-transgenic mouse system using otherwise MHC class I-deficient C57BL/6 mice, thereby conditionally ablating MHC class I-restricted Ag presentation in targeted APC subpopulations. We observed that CD11c+ APCs are critical for early priming of CD8 T cells against the immunodominant TMEV peptide VP2121-130 Loss of H-2Db on CD11c+ APCs mitigates the CD8 T cell response, preventing early viral clearance and immunopathology associated with CD8 T cell activity in the CNS. In contrast, animals with H-2Db-deficient LysM+ APCs retained early priming of Db:VP2121-130 epitope-specific CD8 T cells, although a modest reduction in immune cell entry into the CNS was observed. This work establishes a model enabling the critical dissection of H-2Db-restricted Ag presentation to CD8 T cells, revealing cell-specific and temporal features involved in the generation of CD8 T cell responses. Employing this novel system, we establish CD11c+ cells as pivotal to the establishment of acute antiviral CD8 T cell responses against the TMEV immunodominant epitope VP2121-130, with functional implications both for T cell-mediated viral control and immunopathology.
Collapse
Affiliation(s)
- Zachariah P Tritz
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Robin C Orozco
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Courtney S Malo
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Cori E Fain
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Roman H Khadka
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Emma N Goddery
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Lila T Yokanovich
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905.,Mayo Clinic Department of Immunology, Rochester, MN 55905
| | - Megan L Settell
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905
| | | | - Fang Jin
- Mayo Clinic Department of Immunology, Rochester, MN 55905
| | | | - Larry R Pease
- Mayo Clinic Department of Immunology, Rochester, MN 55905.,Mayo Clinic Department of Biochemistry, Rochester, MN 55905
| | - Aaron J Johnson
- Mayo Clinic Department of Immunology, Rochester, MN 55905; .,Mayo Clinic Department of Molecular Medicine, Rochester, MN 55905; and.,Mayo Clinic Department of Neurology, Rochester, MN 55905
| |
Collapse
|
4
|
Jin YH, Kim CX, Huang J, Kim BS. Infection and Activation of B Cells by Theiler's Murine Encephalomyelitis Virus (TMEV) Leads to Autoantibody Production in an Infectious Model of Multiple Sclerosis. Cells 2020; 9:cells9081787. [PMID: 32727036 PMCID: PMC7465974 DOI: 10.3390/cells9081787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/23/2022] Open
Abstract
Theiler’s murine encephalomyelitis virus (TMEV) induces immune-mediated inflammatory demyelinating disease in susceptible mice that is similar to human multiple sclerosis (MS). In light of anti-CD20 therapies for MS, the susceptibility of B cells to TMEV infection is particularly important. In our study, direct viral exposure to macrophages and lymphocytes resulted in viral replication and cellular stimulation in the order of DCs, macrophages, B cells, and T cells. Notably, B cells produced viral proteins and expressed elevated levels of CD69, an activation marker. Similarly, the expression of major histocompatibility complex class II and costimulatory molecules in B cells was upregulated. Moreover, TMEV-infected B cells showed elevated levels of antigen-presenting function and antibody production. TMEV infection appeared to polyclonally activate B cells to produce autoantibodies and further T cell stimulation. Thus, the viral infection might potentially affect the outcome of autoimmune diseases, and/or the development of other chronic infections, including the protection and/or pathogenesis of TMEV-induced demyelinating disease.
Collapse
Affiliation(s)
- Young-Hee Jin
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Korea
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
- Correspondence: (Y.-H.J.); (B.S.K.); Tel.: +82-42-610-8850 (Y.-H.J.); +1-312-503-8693 (B.S.K.)
| | - Charles X. Kim
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- M Health Fairview Heart Clinic, University of Minnesota Health, Edina, MN 55435, USA
| | - Jocelin Huang
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- M Health Cancer Care, University of Minnesota Health, Edina, MN 55435, USA
| | - Byung S. Kim
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- Correspondence: (Y.-H.J.); (B.S.K.); Tel.: +82-42-610-8850 (Y.-H.J.); +1-312-503-8693 (B.S.K.)
| |
Collapse
|
5
|
Perwein MK, Smestad JA, Warrington AE, Heider RM, Kaczor MW, Maher LJ, Wootla B, Kunbaz A, Rodriguez M. A comparison of human natural monoclonal antibodies and aptamer conjugates for promotion of CNS remyelination: where are we now and what comes next? Expert Opin Biol Ther 2018; 18:545-560. [PMID: 29460650 DOI: 10.1080/14712598.2018.1441284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic and progressive inflammatory demyelinating disease of the human central nervous system (CNS) and is the most common disabling neurological condition in young adults, resulting in severe neurological defects. No curative or long-term progression-inhibiting therapy has yet been developed. However, recent investigation has revealed potential strategies that do not merely modulate potentially pathogenic autoimmune responses, but stimulate remyelination within CNS lesions. AREAS COVERED We discuss the history and development of natural human IgM-isotype immunoglobulins (HIgMs) and recently-identified aptamer-conjugates that have been shown to enhance endogenous myelin repair in animal models of demyelination by acting on myelin-producing oligodendrocytes (OLs) or oligodendrocyte progenitor cells (OPCs) within CNS lesions. We also discuss future development aims and applications for these important novel technologies. EXPERT OPINION Aptamer conjugate Myaptavin-3064 and recombinant human IgM-isotype antibody rHIgM22 regenerate CNS myelin, thereby reducing axonal degeneration and offering the potential of recovery from MS relapses, reversal of disability and prevention of disease progression. Advancement of these technologies into the clinic for MS treatment is therefore a top priority. It remains unclear to what extent the therapeutic modalities of remyelinating antibodies and aptamers may synergize with other currently-approved therapies to yield enhanced therapeutic effects.
Collapse
Affiliation(s)
- Maria K Perwein
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - John A Smestad
- b Medical Scientist Training Program , Mayo Clinic College of Medicine and Science , Rochester , MN , USA.,c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Arthur E Warrington
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Robin M Heider
- c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Mark W Kaczor
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Louis J Maher
- c Department of Biochemistry and Molecular Biology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Bharath Wootla
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Ahmad Kunbaz
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Moses Rodriguez
- a Department of Neurology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA.,d Department of Immunology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
6
|
Wootla B, Denic A, Watzlawik JO, Warrington AE, Zoecklein LJ, Papke-Norton LM, David C, Rodriguez M. Human class I major histocompatibility complex alleles determine central nervous system injury versus repair. J Neuroinflammation 2016; 13:293. [PMID: 27855706 PMCID: PMC5112886 DOI: 10.1186/s12974-016-0759-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hβ2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, β2m0) and class II-deficient (mouse Aβ0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS Following infection with TMEV, a picornavirus, the Aβ0.β2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hβ2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.
Collapse
Affiliation(s)
- Bharath Wootla
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Center for Regenerative Medicine, Neuroregeneration, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Aleksandar Denic
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Jens O. Watzlawik
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road S, Jacksonville, FL 32224 USA
| | - Arthur E. Warrington
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Laurie J. Zoecklein
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Louisa M. Papke-Norton
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Chella David
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
7
|
Rodriguez M, Pease LR, David CS. Immune-mediated injury of virus-infected oligodendrocytes A model of multiple sclerosis. ACTA ACUST UNITED AC 2014; 7:359-63. [PMID: 25291330 DOI: 10.1016/0167-5699(86)90025-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The causes of primary demyelination in diseases such as multiple sclerosis are still unknown, but it is possible that immune attack triggered by virus infection may be responsible. Theiler's murine encephalitis is a popular animal model of demyelinating diseases, and in this article Moses Rodriguez and his colleagues describe a hypothetical scheme to explain differential susceptibility of inbred strains to infection. They propose a mechanism by which specific demyelination is produced when immune cells interact with viral antigen and major histocompatibility complex (MHC) antigens.
Collapse
Affiliation(s)
- M Rodriguez
- Departments of Neurology and Immunology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
8
|
Takizawa S, Kaneyama T, Tsugane S, Takeichi N, Yanagisawa S, Ichikawa M, Yagita H, Kim BS, Koh CS. Role of the Programmed Death-1 (PD-1) pathway in regulation of Theiler's murine encephalomyelitis virus-induced demyelinating disease. J Neuroimmunol 2014; 274:78-85. [PMID: 25027060 DOI: 10.1016/j.jneuroim.2014.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/10/2014] [Accepted: 06/19/2014] [Indexed: 12/28/2022]
Abstract
Programmed death-1 (PD-1) belongs to the CD28 family of co-stimulatory and co-inhibitory molecules and regulates adaptive immunity. This molecule induces the development of regulatory T cells, T cell tolerance, or apoptosis. We examined the role of PD-1 pathway in Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) mice. Up-regulation of PD-1 and PD-1 ligand-1 (PD-L1) mRNA expression in bone marrow-derived dendritic cells were induced by TMEV infection in vitro. Furthermore, PD-1 and PD-L1 mRNA expression was increased in the spinal cords of the TMEV-infected mice in vivo. Treatment with a blocking monoclonal antibody (mAb) against PD-1, especially during the effector phase, resulted in significant deterioration of the TMEV-IDD both clinically and histologically. Flow cytometric analysis revealed a dramatically increase of CD4(+) T cells producing Th1 cytokines such as IFN-γ and TNF-α in the spinal cord of anti-PD-1 mAb-treated mice. These results indicate that the PD-1 pathway plays a pivotal regulatory role in the development of TMEV-IDD.
Collapse
Affiliation(s)
- Sho Takizawa
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Tomoki Kaneyama
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Sayaka Tsugane
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Naoya Takeichi
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Satoshi Yanagisawa
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Motoki Ichikawa
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Byung S Kim
- Department of Microbiology-Immunology, Northwestern University Finberg Medical School, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Chang-Sung Koh
- Department of Biomedical Laboratory Sciences, Graduate School of Medicine, Shinshu University, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
9
|
Pierce M, Rodriguez M. Erichrome Stain for Myelin on Osmicated Tissue Embedded in Glycol Methacrylate Plastic. J Histotechnol 2013. [DOI: 10.1179/his.1989.12.1.35] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
10
|
Modeling the acute and chronic phases of Theiler murine encephalomyelitis virus infection. J Virol 2013; 87:4052-9. [PMID: 23365440 DOI: 10.1128/jvi.03395-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Theiler murine encephalomyelitis virus (TMEV) infection of a mouse's central nervous system is biphasic: first the virus infects motor neurons (acute phase), and this is followed by a chronic phase in which the virus infects glial cells (primarily microglia and macrophages [M]) of the spinal cord white matter, leading to inflammation and demyelination. As such, TMEV-induced demyelinating disease in mice provides a highly relevant experimental animal model for multiple sclerosis. Mathematical models have proven valuable in understanding the in vivo dynamics of persistent virus infections, such as HIV-1, hepatitis B virus, and hepatitis C virus infections. However, viral dynamic modeling has not been used for understanding TMEV infection. We constructed the first mathematical model of TMEV-host kinetics during acute and early chronic infections in mice and fit measured viral kinetic data with the model. The data fitting allowed us to estimate several unknown parameters, including the following: the rate of infection of neurons, 0.5 × 10(-8) to 5.6 × 10(-8) day(-1); the percent reduction of the infection rate due to the presence of virus-specific antibodies, which reaches 98.5 to 99.9% after day 15 postinfection (p.i.); the half-life of infected neurons, 0.1 to 1.2 days; and a cytokine-enhanced macrophage source rate of 25 to 350 M/day into the spinal cord starting at 10.9 to 12.9 days p.i. The model presented here is a first step toward building a comprehensive model for TMEV-induced demyelinating disease. Moreover, the model can serve as an important tool in understanding TMEV infectious mechanisms and may prove useful in evaluating antivirals and/or therapeutic modalities to prevent or inhibit demyelination.
Collapse
|
11
|
Myoung J, Kang HS, Hou W, Meng L, Dal Canto MC, Kim BS. Epitope-specific CD8+ T cells play a differential pathogenic role in the development of a viral disease model for multiple sclerosis. J Virol 2012; 86:13717-28. [PMID: 23055563 PMCID: PMC3503089 DOI: 10.1128/jvi.01733-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/03/2012] [Indexed: 01/01/2023] Open
Abstract
Theiler's virus-induced demyelinating disease has been extensively investigated as a model for persistent viral infection and multiple sclerosis (MS). However, the role of CD8(+) T cells in the development of disease remains unclear. To assess the role of virus-specific CD8(+) T cells in the pathogenesis of demyelinating disease, a single amino acid substitution was introduced into the predominant viral epitope (VP3 from residues 159 to 166 [VP3(159-166)]) and/or a subdominant viral epitope (VP3(173-181)) of susceptible SJL/J mice by site-directed mutagenesis. The resulting variant viruses (N160V, P179A, and N160V/P179A) failed to induce CD8(+) T cell responses to the respective epitopes. Surprisingly, mice infected with N160V or N160V/P179A virus, which lacks CD8(+) T cells against VP3(159-166), did not develop demyelinating disease, in contrast to wild-type virus or P179A virus lacking VP3(173-181)-specific CD8(+) T cells. Our findings clearly show that the presence of VP3(159-166)-specific CD8(+) T cells, rather than viral persistence itself, is strongly correlated with disease development. VP3(173-181)-specific CD8(+) T cells in the central nervous system (CNS) of these virus-infected mice expressed higher levels of transforming growth factor β, forkhead box P3, interleukin-22 (IL-22), and IL-17 mRNA but caused minimal cytotoxicity compared to that caused by VP3(159-166)-specific CD8(+) T cells. VP3(159-166)-specific CD8(+) T cells exhibited high functional avidity for gamma interferon production, whereas VP3(173-181)-specific CD8(+) T cells showed low avidity. To our knowledge, this is the first report indicating that the induction of the IL-17-producing CD8(+) T cell type is largely epitope specific and that this specificity apparently plays a differential role in the pathogenicity of virus-induced demyelinating disease. These results strongly advocate for the careful consideration of CD8(+) T cell-mediated intervention of virus-induced inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | | | - Mauro C. Dal Canto
- Department of Pathology, Northwestern University Medical School, Chicago, Illinois, USA
| | - Byung S. Kim
- Department of Microbiology-Immunology
- Department of Pathology, Northwestern University Medical School, Chicago, Illinois, USA
| |
Collapse
|
12
|
Denic A, Zoecklein L, Kerkvliet J, Papke L, Edukulla R, Warrington A, Bieber A, Pease LR, David CS, Rodriguez M. Transgenic expression of viral capsid proteins predisposes to axonal injury in a murine model of multiple sclerosis. Brain Pathol 2011; 21:501-15. [PMID: 21314744 DOI: 10.1111/j.1750-3639.2011.00474.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We used transgenic expression of capsid antigens to Theiler's murine encephalomyelitis virus (TMEV) to study the influence of VP1, VP2 or VP2(121-130) to either protection or pathogenesis to chronic spinal cord demyelination, axonal loss and functional deficits during the acute and chronic phases of infection. We used both mice that are normally susceptible (FVB) and mice normally resistant (FVB.D(b) ) to demyelination. Transgenic expression of VP2(121-130) epitope in resistant FVB.D(b) mice caused spinal cord pathology and virus persistence because the VP2(121-130) epitope is the dominant peptide recognized by D(b) , which is critical for virus clearance. In contrast, all three FVB TMEV transgenic mice showed more demyelination, inflammation and axonal loss as compared with wild-type FVB mice, even though virus load was not increased. Motor function measured by rotarod showed weak correlation with total number of midthoracic axons, but a strong correlation with large-caliber axons (>10µm(2) ). This study supports the hypothesis that expression of viral capsid proteins as self influences the extent of axonal pathology following Theiler's virus-induced demyelination. The findings provide insight into the role of axonal injury in the development of functional deficits that may have relevance to human demyelinating disease.
Collapse
|
13
|
Yanagisawa S, Takeichi N, Kaneyama T, Yagita H, Taniguchi S, Kim BS, Koh CS. Effects of anti-CD70 mAb on Theiler's murine encephalomyelitis virus-induced demyelinating disease. Brain Res 2010; 1317:236-45. [PMID: 20045400 DOI: 10.1016/j.brainres.2009.12.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 12/17/2009] [Accepted: 12/18/2009] [Indexed: 10/20/2022]
Abstract
Ligation of CD27, a member of the tumor necrosis factor (TNF) receptor family, by its ligand CD70 is thought to be important in T cell activation, expansion and survival, B cell activation, and NK cell activation. We examined the role of CD70 in Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) mice. Blocking of CD70 in effector phase by anti-CD70 monoclonal antibody (mAb) suppressed the development of TMEV-IDD. The number of IFN-gamma- or TNF-alpha-producing cells in the spleen and mRNA levels of IFN-gamma and TNF-alpha in spinal cord were decreased in mice treated with anti-CD70 mAb at the effector phase. In contrast, treatment with anti-CD70 mAb in induction phase failed to reduce these responses, compared to nonspecific IgG-treated control mice. These data suggest that CD70 is critically involved in the pathogenesis of TMEV-IDD and that antibodies against CD70 could be a novel therapeutic approach in the clinical treatment of demyelinating diseases such as human multiple sclerosis.
Collapse
Affiliation(s)
- Satoshi Yanagisawa
- Department of Biomedical Laboratory Sciences, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| | | | | | | | | | | | | |
Collapse
|
14
|
Steelman AJ, Alford E, Young CR, Welsh TH, Meagher MW, Welsh CJR. Restraint stress fails to render C57BL/6 mice susceptible to Theiler's virus-induced demyelination. Neuroimmunomodulation 2010; 17:109-19. [PMID: 19923856 PMCID: PMC3214847 DOI: 10.1159/000258694] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 07/16/2009] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Multiple sclerosis is a degenerative disease of the CNS with a pathology consistent with immunological mediation. Although its cause is unknown, multiple factors are thought to influence both the onset and exacerbation of the disease, including both genetic background as well as environmental factors. METHODS We are interested in the effect of psychological stress on the onset and exacerbation of Theiler's virus-induced demyelinating disease (TVID), a murine model of MS in which viral persistence facilitates demyelination. In the current study, we determined whether chronic restraint stress (RS)-induced immunosuppression could result in the establishment of a persistent CNS infection in the normally TVID-resistant C57BL/6 mouse strain, resulting in demyelination. RESULTS Our data indicated that RS repeated over the course of 7 days was not sufficient to cause decreases in virus-specific adaptive immunity, and did not significantly alter CNS viral levels. Furthermore, chronic repeated RS lasting until 4 weeks after infection altered neither the development of virus-specific IgG nor motor function determined by Rotarod analysis. In addition, histological analysis of the CNS of stressed mice indicated no inflammation or demyelination on day 193 after infection. CONCLUSION These results suggest that stress alone is not sufficient to overcome genetic resistance to TVID in the C57BL/6 mouse strain.
Collapse
MESH Headings
- Adaptive Immunity/immunology
- Animals
- Cardiovirus Infections/immunology
- Cardiovirus Infections/psychology
- Central Nervous System/immunology
- Central Nervous System/pathology
- Central Nervous System/virology
- Demyelinating Autoimmune Diseases, CNS/immunology
- Demyelinating Autoimmune Diseases, CNS/physiopathology
- Demyelinating Autoimmune Diseases, CNS/psychology
- Disease Models, Animal
- Disease Susceptibility/immunology
- Disease Susceptibility/psychology
- Female
- Genetic Predisposition to Disease/genetics
- Immune Tolerance/genetics
- Immune Tolerance/immunology
- Mice
- Mice, Inbred C57BL
- Movement Disorders/immunology
- Movement Disorders/physiopathology
- Nerve Fibers, Myelinated/immunology
- Nerve Fibers, Myelinated/pathology
- Nerve Fibers, Myelinated/virology
- Neurons/immunology
- Neurons/pathology
- Neurons/virology
- Restraint, Physical/adverse effects
- Restraint, Physical/psychology
- Stress, Psychological/immunology
- Theilovirus/immunology
- Viral Load/immunology
Collapse
Affiliation(s)
- Andrew J. Steelman
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Tex., USA
| | - Eric Alford
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Tex., USA
| | - Colin R. Young
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Tex., USA
| | - Thomas H. Welsh
- Department of Animal Science, College of Agriculture, Texas A&M University, College Station, Tex., USA
| | - Mary W. Meagher
- Department of Psychology, College of Liberal Arts, Texas A&M University, College Station, Tex., USA
| | - C. Jane R. Welsh
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Tex., USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Tex., USA
- *Dr. C. Jane Welsh, Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458 (USA), Tel. +1 979 862 4974, Fax +1 979 847 8981, E-Mail
| |
Collapse
|
15
|
Anticapsid immunity level, not viral persistence level, correlates with the progression of Theiler's virus-induced demyelinating disease in viral P1-transgenic mice. J Virol 2008; 82:5606-17. [PMID: 18353953 DOI: 10.1128/jvi.02442-07] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intracranial infection of Theiler's murine encephalomyelitis virus (TMEV) induces demyelination and a neurological disease in susceptible SJL/J (SJL) mice that resembles multiple sclerosis. While the virus is cleared from the central nervous system (CNS) of resistant C57BL/6 (B6) mice, it persists in SJL mice. To investigate the role of viral persistence and its accompanying immune responses in the development of demyelinating disease, transgenic mice expressing the P1 region of the TMEV genome (P1-Tg) were employed. Interestingly, P1-Tg mice with the B6 background showed severe reductions in both CD4(+) and CD8(+) T-cell responses to capsid epitopes, while P1-Tg mice with the SJL background displayed transient reductions following viral infection. Reduced antiviral immune responses in P1-Tg mice led to >100- to 1,000-fold increases in viral persistence at 120 days postinfection in the CNS of mice with both backgrounds. Despite the increased CNS TMEV levels in these P1-Tg mice, B6 P1-Tg mice developed neither neuropathological symptoms nor demyelinating lesions, and SJL P1-Tg mice developed significantly less severe TMEV-induced demyelinating disease. These results strongly suggest that viral persistence alone is not sufficient to induce disease and that the level of T-cell immunity to viral capsid epitopes is critical for the development of demyelinating disease in SJL mice.
Collapse
|
16
|
Jin YH, Mohindru M, Kang MH, Fuller AC, Kang B, Gallo D, Kim BS. Differential virus replication, cytokine production, and antigen-presenting function by microglia from susceptible and resistant mice infected with Theiler's virus. J Virol 2007; 81:11690-702. [PMID: 17715222 PMCID: PMC2168808 DOI: 10.1128/jvi.01034-07] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infection with Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) causes an immune system-mediated demyelinating disease similar to human multiple sclerosis in susceptible but not resistant strains of mice. To understand the underlying mechanisms of differential susceptibility, we analyzed viral replication, cytokine production, and costimulatory molecule expression levels in microglia and macrophages in the CNS of virus-infected resistant C57BL/6 (B6) and susceptible SJL/J (SJL) mice. Our results indicated that message levels of TMEV, tumor necrosis factor alpha, beta interferon, and interleukin-6 were consistently higher in microglia from virus-infected SJL mice than in those from B6 mice. However, the levels of costimulatory molecule expression, as well as the ability to stimulate allogeneic T cells, were significantly lower in TMEV-infected SJL mice than in B6 mice. In addition, microglia from uninfected naïve mice displayed differential viral replication, T-cell stimulation, and cytokine production, similar to those of microglia from infected mice. These results strongly suggest that different levels of intrinsic susceptibility to TMEV infection, cytokine production, and T-cell activation ability by microglia contribute to the levels of viral persistence and antiviral T-cell responses in the CNS, which are critical for the differential susceptibility to TMEV-induced demyelinating disease between SJL and B6 mice.
Collapse
Affiliation(s)
- Young-Hee Jin
- Department of Microbiology and Immunology, Northwestern University Feinberg Medical School, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Mohindru M, Kang B, Kim BS. Initial capsid-specific CD4(+) T cell responses protect against Theiler's murine encephalomyelitisvirus-induced demyelinating disease. Eur J Immunol 2006; 36:2106-15. [PMID: 16761311 DOI: 10.1002/eji.200535785] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Central nervous system (CNS) infection by Theiler's murine encephalomyelitis virus (TMEV) causes an immune-mediated demyelinating disease similar to human multiple sclerosis in susceptible mice. To understand the pathogenic mechanisms, we analyzed the level, specificity, and function of CD4(+) Th cells in susceptible SJL/J and resistant C57BL/6 mice. Compared to resistant mice, susceptible mice have three- to fourfold higher levels of overall CNS-infiltrating CD4(+) T cells during acute infection. CD4(+) T cells in the CNS of both strains display various activation markers and produce high levels of IFN-gamma upon stimulation with anti-CD3 antibody. However, susceptible mice display significantly fewer (tenfold) IFN-gamma-producing Th1 cells specific for viral capsid epitopes as compared to resistant mice. Furthermore, preimmunization with capsid-epitope peptides significantly increased capsid-specific CD4(+) T cells in the CNS during the early stages of viral infection and delayed the development of demyelinating disease in SJL/J mice. This suggests a protective role of capsid-reactive Th cells during early viral infection. Therefore, a low level of the protective Th1 response to viral capsid proteins, in conjunction with Th1 responses to unknown epitopes may delay viral clearance in susceptible mice leading to pathogenesis of demyelination during acute infection, as compared to resistant mice.
Collapse
Affiliation(s)
- Mani Mohindru
- Department of Microbiology-Immunology, Northwestern University Feinberg Medical School, Chicago, IL 60611, USA
| | | | | |
Collapse
|
18
|
So EY, Kang MH, Kim BS. Induction of chemokine and cytokine genes in astrocytes following infection with Theiler's murine encephalomyelitis virus is mediated by the Toll-like receptor 3. Glia 2006; 53:858-67. [PMID: 16586493 DOI: 10.1002/glia.20346] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection in the central nervous system (CNS) induces a demyelinating disease similar to human multiple sclerosis. TMEV infection results in activation of various chemokine and cytokine genes that are important in the initiation of an inflammatory response. We have previously shown that the production of these chemokines and cytokines in astrocytes is induced via the NF-kappaB pathway following TMEV and Coxsackie virus infection. In this study, we investigated whether the NF-kappaB-dependent inflammatory responses after TMEV infection is triggered through TLR3 and/or TLR7. The activation of NF-kappaB or IRF/ISRE, as well as the production of both MCP-1/CCL2 and IL-8/CXCL8, was observed in only TLR3-transfected HEK 293 cells, but not in TLR7-tranfected cells. The potential involvement of TLR3 in mouse embryonic fibroblasts and primary astrocytes was further investigated following transfection with wildtype or dominant negative form of TLRs and MyD88, as well as astrocytes from TLR3- and MyD88-deficient mice. Similarly, the activation of transcription factors and chemokine genes is induced in these mouse cells through primarily TLR3 signaling pathway, but not TLR7 or other MyD88-mediated pathways following TMEV infection. However, the TLR3-mediated cellular activation does not appear to affect the level of viral replication in astrocytes. These results strongly suggest that TLR3-signaling by TMEV alone is sufficient to induce the initial inflammatory cytokine responses that could be very important for the outcome of virus-induced encephalitis and/or demyelinating diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- Eui Young So
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois, USA
| | | | | |
Collapse
|
19
|
Drescher KM, Murray PD, David CS, Pease LR, Rodriguez M. CNS cell populations are protected from virus-induced pathology by distinct arms of the immune system. Brain Pathol 2006; 9:21-31. [PMID: 9989447 PMCID: PMC8098348 DOI: 10.1111/j.1750-3639.1999.tb00206.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The basis for the distinct patterns of brain pathology in individuals experiencing virus-induced encephalitis may be related to either the tropism of the virus or the host's response to virus infection of the central nervous system (CNS). In these studies we used Theiler's murine encephalomyelitis virus (TMEV) and a series of mice deficient in various immune system components (alpha/beta T cells, antibody, Class I MHC, and Class II MHC) to examine the hypothesis that discrete populations of CNS cells are protected differentially from virus infection by distinct arms of the immune response. Here we demonstrate that the Class I-mediated immune response provided more protection from areas of the brain (brainstem, corpus callosum and cerebellum) with abundant white matter as there was significantly more disease in these areas in beta2m -/- (Class I-deficient) mice as compared to A beta(0) (Class II-deficient) mice. In contrast, the striatum, with an abundance of neurons, was protected from virus-induced pathology primarily by antibody. In addition, we determined that antibody and alpha/beta T cells provided protection from severe deficits and death during the acute phase of the disease. The data presented here support the hypothesis that distinct immune system components function to protect discrete areas of the CNS from virus-induced pathology.
Collapse
Affiliation(s)
- K M Drescher
- Department of Immunology, Mayo Medical School, Rochester, MN 55901, USA
| | | | | | | | | |
Collapse
|
20
|
Drescher KM, Zoecklein LJ, Pavelko KD, Rivera‐Quinones C, Hollenbaugh D, Rodriguez M. CD40L is critical for protection from demyelinating disease and development of spontaneous remyelination in a mouse model of multiple sclerosis. Brain Pathol 2006; 10:1-15. [PMID: 10668891 PMCID: PMC8098531 DOI: 10.1111/j.1750-3639.2000.tb00238.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) induces acute neuronal disease followed by chronic demyelination in susceptible strains of mice. In this study we examined the role of a limited immune defect (deletion or blocking of CD40 ligand [CD40L]) on the extent of brain disease, susceptibility to demyelination, and the ability of demyelinated mice to spontaneously remyelinate following TMEV infection. We demonstrated that CD40L-dependent immune responses participate in pathogenesis in the cerebellum and the spinal cord white matter but protect the striatum of susceptible SJL/J mice. In mice on a background resistant to TMEV-induced demyelination (C57BL/6), the lack of CD40L resulted in increased striatal disease and meningeal inflammation. In addition, CD40L was required to maintain resistance to demyelination and clinical deficits in H-2b mice. CD40L-mediated interactions were also necessary for development of protective H-2b-restricted cytotoxic T cell responses directed against the VP2 region of TMEV as well as for spontaneous remyelination of the spinal cord white matter. The data presented here demonstrated the critical role of this molecule in both antibody- and cell-mediated protective immune responses in distinct phases of TMEV-mediated pathology.
Collapse
Affiliation(s)
- Kristen M. Drescher
- Departments of Immunology and Neurology, Mayo Medical School, Rochester, MN 55905
| | - Laurie J. Zoecklein
- Departments of Immunology and Neurology, Mayo Medical School, Rochester, MN 55905
| | - Kevin D. Pavelko
- Departments of Immunology and Neurology, Mayo Medical School, Rochester, MN 55905
| | | | | | - Moses Rodriguez
- Departments of Immunology and Neurology, Mayo Medical School, Rochester, MN 55905
| |
Collapse
|
21
|
Tsunoda I, Tolley ND, Theil DJ, Whitton JL, Kobayashi H, Fujinami RS. Exacerbation of viral and autoimmune animal models for multiple sclerosis by bacterial DNA. Brain Pathol 2006; 9:481-93. [PMID: 10416988 PMCID: PMC8098503 DOI: 10.1111/j.1750-3639.1999.tb00537.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection and relapsing-remitting experimental allergic encephalomyelitis (R-EAE) have been used to investigate the viral and autoimmune etiology of multiple sclerosis (MS), a possible Th1-type mediated disease. DNA immunization is a novel vaccination strategy in which few harmful effects have been reported. Bacterial DNA and oligodeoxynucleotides, which contain CpG motifs, have been reported to enhance immunostimulation. Our objectives were two-fold: first, to ascertain whether plasmid DNA, pCMV, which is widely used as a vector in DNA immunization studies, could exert immunostimulation in vitro; and second, to test if pCMV injection could modulate animal models for MS in vivo. We demonstrated that this bacterially derived DNA could induce interleukin (IL)-12, interferon (IFN)gamma, (Th1-promoting cytokines), and IL-6 production as well as activate NK cells. Following pCMV injections, SJL/J mice were infected with TMEV or challenged with encephalitogenic myelin proteolipid protein (PLP) peptides. pCMV injection exacerbated TMEV-induced demyelinating disease in a dose-dependent manner. Exacerbation of the disease did not correlate with the number of TMEV-antigen positive cells but did with an increase in anti-TMEV antibody. pCMV injection also enhanced R-EAE with increased IFNgamma and IL-6 responses. These results caution the use of DNA vaccination in MS patients and other possible Th1-mediated diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- CpG Islands/immunology
- Cytokines/biosynthesis
- Cytomegalovirus/immunology
- DNA, Bacterial/adverse effects
- DNA, Bacterial/immunology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Genetic Vectors/adverse effects
- Genetic Vectors/immunology
- Immunoglobulin G/blood
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred Strains
- Multiple Sclerosis/immunology
- Multiple Sclerosis/microbiology
- Multiple Sclerosis/pathology
- Myelin Proteolipid Protein/immunology
- Spinal Cord/pathology
- Th1 Cells/immunology
- Theilovirus/immunology
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- I Tsunoda
- Department of Neurology, University of Utah School of Medicine, Salt Lake City 84132, USA
| | | | | | | | | | | |
Collapse
|
22
|
Grigoriadis N, Hadjigeorgiou GM. Virus-mediated autoimmunity in Multiple Sclerosis. JOURNAL OF AUTOIMMUNE DISEASES 2006; 3:1. [PMID: 16504001 PMCID: PMC1397830 DOI: 10.1186/1740-2557-3-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2005] [Accepted: 02/19/2006] [Indexed: 11/21/2022]
Abstract
Epidemiological data suggest the notion that in Multiple Sclerosis (MS) is an acquired autoimmune disease and the cause may be an environmental factor(s), probably infectious, in genetically susceptible individuals. Several cases of viral induced demyelinatimg encephalomyelitis in human beings and in experimental models as well as the presence of IgG oligoclonal bands in the cerebrospinal fluid indicate that the infectious factor may be viral. However, the absence of a specific virus identification in MS central nervous system may hardly support this notion. On the other hand, the partial response of patients with MS to immunosuppressive and immunomodulatory therapy support the evidence of an autoimmune etiology for MS. However, the autoimmune hypothesis shares the same criticism with the infectious one in that no autoantigen(s) specific to and causative for MS has ever been identified. Nevertheless, the absence of identifiable infectious agent, especially viral does not rule out its presence at a certain time – point and the concomitant long term triggering of an autoimmune cascade of events thereafter. Several concepts have emerged in an attempt to explain the autoimmune mechanisms and ongoing neurodegeneration in MS on the basis of the infectious – viral hypothesis.
Collapse
Affiliation(s)
- Nikolaos Grigoriadis
- B' Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, 1 Stilp Kyriakidi Street, Aristotle University of Thessaloniki, Thessaloniki, 54636 Thessaloniki, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, Neurogenetics Unit, Medical School, University of Thessaly, 22 Papakyriazi Street, 41222 Larissa, Greece
| |
Collapse
|
23
|
Kang B, Kang HK, Kim BS. Identification of capsid epitopes of Theiler's virus recognized by CNS-infiltrating CD4+ T cells from virus-infected C57BL/6 mice. Virus Res 2005; 108:57-61. [PMID: 15681055 DOI: 10.1016/j.virusres.2004.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2004] [Revised: 08/03/2004] [Accepted: 08/03/2004] [Indexed: 11/19/2022]
Abstract
Intracerebral infection of Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated demyelinating disease in some mouse strains but not in others. We report here for the first time two new predominant capsid epitopes (VP4(21-40) and VP2(201-220)) recognized by CD4+ T cells from virus-infected resistant C57BL/6 mice based on IFNgamma ELISPOT assay utilizing a 20-mer peptide library covering the entire capsid proteins. Further experiments by IFNgamma ELISPOT and flow cytometry for intracellular IFNgamma production using truncated peptides indicated that the epitope regions recognized by CNS-infiltrating CD4+ T cells are VP4(25-38) and VP2(206-220), respectively. No apparent reduction in the T cell response to these viral epitopes is seen in the CNS of IL-12- and ICAM-1-deficient C57BL/6 mice compared to those in control C57BL/6 mice, suggesting that T cell response to TMEV in the CNS is largely insensitive to the absence of these proinflammatory cytokine and adhesion molecules. Therefore, these newly defined CD4+ T cell epitopes are likely to provide an important tool to investigate the role of CD4+ T cell responses in H-2b-bearing congenic strains.
Collapse
Affiliation(s)
- Bongsu Kang
- Department of Microbiology-Immunology, Northwestern University Feinberg Medical School, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | | | |
Collapse
|
24
|
Kim BS, Palma JP, Kwon D, Fuller AC. Innate immune response induced by Theiler's murine encephalomyelitis virus infection. Immunol Res 2005; 31:1-12. [PMID: 15591618 PMCID: PMC7090574 DOI: 10.1385/ir:31:1:01] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although the causative agents of human multiple sclerosis (MS) are not known, it is suspected that a viral infection may be associated with the initiation of the disease. Several viral disease models in mice have been studied to understand the pathogenesis of demeylination. In particular, Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) has been extensively studied as a relevant model. Various cytokines and chemokines are produced upon viral infection by different cell types, including antigen-presenting cells (APCs) such as macrophages; dendritic cells (DCs); and glial cells, such as astrocytes, microglia, and oligoden-drocytes. The upregulation of the corresponding molecules are also found in MS and are likely to play an important role in the protection and/or pathogenesis of chronic inflammatory demyelinating disease. In this review, the type of cells and molecules, gene-activation mechanisms as well as their potential roles in protection and pathogenesis will be discussed.
Collapse
Affiliation(s)
- Byung S Kim
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, IL 60611, USA.
| | | | | | | |
Collapse
|
25
|
Kumar ASM, Reddi HV, Kung AY, Dal Canto M, Lipton HL. Virus persistence in an animal model of multiple sclerosis requires virion attachment to sialic acid coreceptors. J Virol 2004; 78:8860-7. [PMID: 15280494 PMCID: PMC479047 DOI: 10.1128/jvi.78.16.8860-8867.2004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Persistent Theiler's virus infection in the central nervous system (CNS) of mice provides a highly relevant animal model for multiple sclerosis. The low-neurovirulence DA strain uses sialic acid as a coreceptor for cell binding before establishing infection. During adaptation of DA virus to growth in sialic acid-deficient cells, three amino acid substitutions (G1100D, T1081I, and T3182A) in the capsid arose, and the virus no longer used sialic acid as a coreceptor. The adapted virus retained acute CNS virulence, but its persistence in the CNS, white matter inflammation, and demyelination were largely abrogated. Infection of murine macrophage but not oligodendrocyte cultures with the adapted virus was also significantly reduced. Substitution of G1100D in an infectious DA virus cDNA clone demonstrated a major role for this mutation in loss of sialic acid binding and CNS persistence. These data indicate a direct role for sialic acid binding in Theiler's murine encephalomyelitis virus persistence and chronic demyelinating disease.
Collapse
Affiliation(s)
- A S Manoj Kumar
- Department of Neurology, Evanston Hospital, 2650 Ridge Ave., Evanston, IL 60201, USA
| | | | | | | | | |
Collapse
|
26
|
Oleszak EL, Chang JR, Friedman H, Katsetos CD, Platsoucas CD. Theiler's virus infection: a model for multiple sclerosis. Clin Microbiol Rev 2004; 17:174-207. [PMID: 14726460 PMCID: PMC321460 DOI: 10.1128/cmr.17.1.174-207.2004] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Both genetic background and environmental factors, very probably viruses, appear to play a role in the etiology of multiple sclerosis (MS). Lessons from viral experimental models suggest that many different viruses may trigger inflammatory demyelinating diseases resembling MS. Theiler's virus, a picornavirus, induces in susceptible strains of mice early acute disease resembling encephalomyelitis followed by late chronic demyelinating disease, which is one of the best, if not the best, animal model for MS. During early acute disease the virus replicates in gray matter of the central nervous system but is eliminated to very low titers 2 weeks postinfection. Late chronic demyelinating disease becomes clinically apparent approximately 2 weeks later and is characterized by extensive demyelinating lesions and mononuclear cell infiltrates, progressive spinal cord atrophy, and axonal loss. Myelin damage is immunologically mediated, but it is not clear whether it is due to molecular mimicry or epitope spreading. Cytokines, nitric oxide/reactive nitrogen species, and costimulatory molecules are involved in the pathogenesis of both diseases. Close similarities between Theiler's virus-induced demyelinating disease in mice and MS in humans, include the following: major histocompatibility complex-dependent susceptibility; substantial similarities in neuropathology, including axonal damage and remyelination; and paucity of T-cell apoptosis in demyelinating disease. Both diseases are immunologically mediated. These common features emphasize the close similarities of Theiler's virus-induced demyelinating disease in mice and MS in humans.
Collapse
Affiliation(s)
- Emilia L Oleszak
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19106, USA.
| | | | | | | | | |
Collapse
|
27
|
Experimental Models of Virus-Induced Demyelination. MYELIN BIOLOGY AND DISORDERS 2004. [PMCID: PMC7155523 DOI: 10.1016/b978-012439510-7/50097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This chapter reviews two of the most widely studied animal models of virus-induced demyelinating disease. These are Theiler's murine encephalomyelitis virus and murine hepatitis virus. Both viruses produce acute inflammatory encephalitis that is followed by chronic central-nervous-system (CNS) demyelinating disease. The clinical and pathologic correlates of virus-induced demyelination are largely immune mediated. Furthermore, several pathologic mechanisms have been proposed to explain the development of myelin damage and neurologic deficits, and each of the proposed mechanisms may play a role in disease progression depending on the genetic constitution of the infected animal. The induction of demyelinating disease by virus may be directly relevant to human MS. Several viruses are known to cause demyelination in humans and viral infection is an epidemiologic factor that is consistently associated with clinical exacerbation of MS. It is suggested that viral infection may be a cause of MS, although no specific virus has been identified as a causative agent.
Collapse
|
28
|
Rodriguez M, Zoecklein LJ, Howe CL, Pavelko KD, Gamez JD, Nakane S, Papke LM. Gamma interferon is critical for neuronal viral clearance and protection in a susceptible mouse strain following early intracranial Theiler's murine encephalomyelitis virus infection. J Virol 2003; 77:12252-65. [PMID: 14581562 PMCID: PMC254254 DOI: 10.1128/jvi.77.22.12252-12265.2003] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We evaluated the role of gamma interferon (IFN-gamma) in protecting neurons from virus-induced injury following central nervous system infection. IFN-gamma(-/-) and IFN-gamma(+/+) mice of the resistant major histocompatibility complex (MHC) H-2(b) haplotype and intracerebrally infected with Theiler's murine encephalomyelitis virus (TMEV) cleared virus infection from anterior horn cell neurons. IFN-gamma(+/+) H-2(b) mice also cleared virus from the spinal cord white matter, whereas IFN-gamma(-/-) H-2(b) mice developed viral persistence in glial cells of the white matter and exhibited associated spinal cord demyelination. In contrast, infection of IFN-gamma(-/-) mice of the susceptible H-2(q) haplotype resulted in frequent deaths and severe neurologic deficits within 16 days of infection compared to the results obtained for controls. Morphologic analysis demonstrated severe injury to spinal cord neurons in IFN-gamma(-/-) H-2(q) mice during early infection. More virus RNA was detected in the brain and spinal cord of IFN-gamma(-/-) H-2(q) mice than in those of control mice at 14 and 21 days after TMEV infection. Virus antigen was localized predominantly to anterior horn cells in infected IFN-gamma(-/-) H-2(q) mice. IFN-gamma deletion did not affect the humoral response directed against the virus. However, the level of expression of CD4, CD8, class I MHC, or class II MHC in the central nervous system of IFN-gamma(-/-) H-2(q) mice was lower than those in IFN-gamma(+/+) H-2(q) mice. Finally, in vitro analysis of virus-induced death in NSC34 cells and spinal motor neurons showed that IFN-gamma exerted a neuroprotective effect in the absence of other aspects of the immune response. These data support the hypothesis that IFN-gamma plays a critical role in protecting spinal cord neurons from persistent infection and death.
Collapse
Affiliation(s)
- Moses Rodriguez
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
We evaluated the role of interleukin-6 (IL-6) in neuronal injury after CNS infection. IL-6-/- and IL-6+/+ mice of resistant major histocompatibility complex (MHC) H-2b haplotype intracerebrally infected with Theiler's virus cleared the infection normally without development of viral persistence, lethal neuronal infection, or late phase demyelination. In contrast, infection of IL-6-/- mice on a susceptible H-2q haplotype resulted in frequent deaths and severe neurologic deficits within 2 weeks of infection as compared with infected IL-6+/+ H-2q littermate controls. Morphologic analysis demonstrated dramatic injury to anterior horn neurons of IL-6-/- H-2q mice at 12 d after infection. Infectious viral titers in the CNS (brain and spinal cord combined) were equivalent between IL-6-/- H-2q and IL-6+/+ H-2q mice. In contrast, more viral RNA was detected in the spinal cord of IL-6-/- mice compared with IL-6+/+ H-2q mice. Virus antigen was localized predominantly to anterior horn cells in infected IL-6-/- H-2q mice. IL-6 deletion did not affect the humoral response directed against virus, nor did it affect the expression of CD4, CD8, MHC class I, or MHC class II in the CNS. Importantly, IL-6 was expressed by astrocytes of infected IL-6+/+ mice but not in astrocytes of IL-6-/- mice or uninfected IL-6+/+ mice. Furthermore, expression of various chemokines was robust at 12 d after infection in both H-2b and H-2q IL-6-/- mice, indicating that intrinsic CNS inflammatory responses did not depend on the presence of IL-6. Finally, in vitro analysis of virus-induced death in neuroblastoma-spinal cord-34 motor neurons and primary anterior horn cell neurons showed that IL-6 exerted a neuroprotective effect. These data support the hypothesis that IL-6 plays a critical role in protecting specific populations of neurons from irreversible injury.
Collapse
|
30
|
Drescher KM, Zoecklein LJ, Rodriguez M. ICAM-1 is crucial for protection from TMEV-induced neuronal damage but not demyelination. J Neurovirol 2002; 8:452-8. [PMID: 12402172 DOI: 10.1080/13550280260422767] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previous work has suggested that the factors protecting mice from Theiler's murine encephalomyelitis virus (TMEV)-induced spinal cord demyelination are distinct from those involved in protection of the brain during the acute encephalitic phase. In this study, we examined the requirement for intercellular adhesion molecule-1 (ICAM-1) in both of these processes. During the acute phase of infection (days 7 to 10 after intracerebral infection with TMEV), no differences in brain or spinal cord pathology or virus burdens were observed between ICAM-1-knockout mice and the infected immunocompetent control mice of a similar background. Examination of brain pathology later in infection (that is, day 45 post infection [p.i.]) revealed that ICAM-1-deficient mice experienced increased levels of pathology in gray matter regions of the brain. We observed an increase in striatal damage and meningeal inflammation in the brains of TMEV-infected ICAM-1-knockout mice compared to C57BL/6J mice. Despite the increase in brain pathology, no immunoreactivity to viral antigens was detected, suggesting that the virus had been cleared by this time. Resistance to demyelination was similar in both groups, indicating that the resulting immune response was sufficient for protection of the spinal cord white matter.
Collapse
Affiliation(s)
- Kristen M Drescher
- Department of Medical Microbiology and Immunology, Creighton University, Omaha, Nebraska, USA
| | | | | |
Collapse
|
31
|
Lin X, Njenga MK, Johnson AJ, Pavelko KD, David CS, Pease LR, Rodriguez M. Transgenic expression of Theiler's murine encephalomyelitis virus genes in H-2(b) mice inhibits resistance to virus-induced demyelination. J Virol 2002; 76:7799-811. [PMID: 12097592 PMCID: PMC136370 DOI: 10.1128/jvi.76.15.7799-7811.2002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the role of the immune system in protecting against virus-induced demyelination by generating lines of transgenic B10 (H-2(b)) congenic mice expressing three independent contiguous coding regions of the Theiler's murine encephalomyelitis virus (TMEV) under the control of a class I major histocompatibility complex (MHC) promoter. TMEV infection of normally resistant B10 mice results in virus clearance and development of inflammatory demyelination in the spinal cord. Transgenic expression of the viral capsid genes resulted in inactivation of virus-specific CD8(+) T lymphocytes (class I MHC immune function) directed against the relevant peptides, but it did not affect production of virus capsid-specific antibodies or lymphocyte proliferation to the virus antigen (class II MHC immune functions). Following intracerebral infection with TMEV, all three lines of mice survived the acute encephalitis but transgenic mice expressing VP1 (or the cluster of virus capsid proteins [VP4, VP2, and VP3] mapping to the left of VP1 in the TMEV genome) developed virus persistence and subsequent demyelination in spinal cord white matter. Transgenic mice expressing noncapsid proteins mapping to the right of VP1 (2A, 2B, 2C, 3A, 3B, 3C, and 3D) cleared the virus and did not develop demyelination. These results are consistent with the hypothesis that virus capsid gene products of TMEV stimulate class I-restricted CD8(+) T-cell immune responses, which are important for virus clearance and for protection against myelin destruction. Presented within the context of self-antigens, inactivation of these cells by ubiquitous expression of relevant virus capsid peptides partially inhibited resistance to virus-induced demyelination.
Collapse
Affiliation(s)
- Xiaoqi Lin
- Department of Neurology, University of Chicago Medical Center, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Begolka WS, Haynes LM, Olson JK, Padilla J, Neville KL, Dal Canto M, Palma J, Kim BS, Miller SD. CD8-deficient SJL mice display enhanced susceptibility to Theiler's virus infection and increased demyelinating pathology. J Neurovirol 2001; 7:409-20. [PMID: 11582513 PMCID: PMC7094986 DOI: 10.1080/135502801753170264] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Theiler's murine encephalomyelitis virus (TMEV) infection of the central nervous system (CNS) induces a chronic, progressive demyelinating disease in susceptible mouse strains characterized by inflammatory mononuclear infiltrates and spastic hind limb paralysis. Our lab has previously demonstrated a critical role for TMEV- and myelin-specific CD4(+) T cells in initiating and perpetuating this pathology. It has however, also been shown that the MHC class I loci are associated with susceptibility/resistance to TMEV infection and persistence. For this reason, we investigated the contribution of CD8(+) T cells to the TMEV-induced demyelinating pathology in the highly susceptible SJL/J mouse strain. Here we show that beta2M-deficient SJL mice have similar disease incidence rates to wild-type controls, however beta2M-deficient mice demonstrated earlier onset of clinical disease, elevated in vitro responses to TMEV and myelin proteolipid (PLP) epitopes, and significantly higher levels of CNS demyelination and macrophage infiltration at 50 days post-infection. beta2M-deficient mice also displayed a significant elevation in persisting viral titers, as well as an increase in macrophage-derived pro-inflammatory cytokine mRNA expression in the spinal cord at this same time point. Taken together, these results indicate that CD8(+) T cells are not required for clinical or histologic disease initiation or progression in TMEV-infected SJL mice. Rather, these data stress the critical role of CD4(+) T cells in this capacity and further emphasize the potential for CD8(+) T cells to contribute to protection from TMEV-induced demyelination.
Collapse
Affiliation(s)
- Wendy Smith Begolka
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Lia M. Haynes
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Julie K. Olson
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Josette Padilla
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Katherine L. Neville
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Mauro Dal Canto
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Joann Palma
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Byung S. Kim
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| | - Stephen D. Miller
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, 60611 Chicago, IL USA
- Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois USA
| |
Collapse
|
33
|
Abstract
To elucidate the early events in Theiler's virus-induced demyelination, a model for human multiple sclerosis (MS), chemokine gene activation in the central nervous system (CNS) resident cells upon viral infection was investigated. Viral infection selectively upregulated RANTES and IP-10 gene expression in primary astrocyte cultures and broader chemokine genes in oligodendrocyte and microglia cultures. Both RANTES and IP-10 were stimulated by proinflammatory cytokine interferon-gamma (IFNgamma), but only RANTES by tumor necrosis factor alpha (TNFalpha), suggesting that virus infection induces chemokines overlapping with those inducible by proinflammatory cytokines. These results suggest that glial cells, astrocytes in particular, may be critical for early recruitment of inflammatory cells in the initiation of virus-induced, immune-mediated demyelination.
Collapse
Affiliation(s)
- J P Palma
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
34
|
Pathogenic Immunity in Theiler’s Virus-Induced Demyelinating Disease: A Viral Model for Multiple Sclerosis. Autoimmunity 2001. [DOI: 10.1007/978-94-010-0981-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Neville KL, Dal Canto MC, Bluestone JA, Miller SD. CD28 costimulatory blockade exacerbates disease severity and accelerates epitope spreading in a virus-induced autoimmune disease. J Virol 2000; 74:8349-57. [PMID: 10954534 PMCID: PMC116345 DOI: 10.1128/jvi.74.18.8349-8357.2000] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) is a natural mouse pathogen which causes a lifelong persistent infection of the central nervous system (CNS) accompanied by T-cell-mediated myelin destruction leading to chronic, progressive hind limb paralysis. TMEV-induced demyelinating disease (TMEV-IDD) is considered to be a highly relevant animal model for the human autoimmune disease multiple sclerosis (MS), which is thought to be initiated as a secondary consequence of a virus infection. Although TMEV-IDD is initiated by virus-specific CD4(+) T cells targeting CNS-persistent virus, CD4(+) T-cell responses against self myelin protein epitopes activated via epitope spreading contribute to chronic disease pathogenesis. We thus examined the ability of antibodies directed against B7 costimulatory molecules to regulate this chronic virus-induced immunopathologic process. Contrary to previous studies showing that blockade of B7-CD28 costimulatory interactions inhibit the initiation of experimental autoimmune encephalomyelitis, treatment of SJL mice at the time of TMEV infection with murine CTLA-4 immunoglobulin or a combination of anti-B7-1 and anti-B7-2 antibodies significantly enhanced clinical disease severity. Costimulatory blockade inhibited early TMEV-specific T-cell and antibody responses critical in clearing peripheral virus infection. The inhibition of virus-specific immune responses led to significantly increased CNS viral titers resulting in increased damage to myelin-producing oligodendrocytes. Following clearance of the costimulatory antagonists, epitope spreading to myelin epitopes was accelerated as a result of the increased availability of myelin epitopes leading to a more severe chronic disease course. Our results raise concern about the potential use of B7-CD28 costimulatory blockade to treat human autoimmune diseases potentially associated with acute or persistent virus infections.
Collapse
Affiliation(s)
- K L Neville
- Department of Microbiology-Immunology, Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
36
|
Pavelko KD, Drescher KM, McGavern DB, David CS, Rodriguez M. HLA-DQ polymorphism influences progression of demyelination and neurologic deficits in a viral model of multiple sclerosis. Mol Cell Neurosci 2000; 15:495-509. [PMID: 10860577 PMCID: PMC5450945 DOI: 10.1006/mcne.2000.0843] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The importance of genetic susceptibility in determining the progression of demyelination and neurologic deficits is a major focus in neuroscience. We studied the influence of human leukocyte antigen (HLA)-DQ polymorphisms on disease course and neurologic impairment in virus-induced demyelination. HLA-DQ6 or DQ8 was inserted as a transgene into mice lacking endogenous expression of MHC class I (beta(2)m) and class II (H2-A(beta)) molecules. Following Theiler's murine encephalomyelitis virus (TMEV) infection, we assessed survival, virus persistence, demyelination, and clinical disease. Mice lacking expression of endogenous class I and class II molecules (beta(2)m(o) Abeta(o) mice) died 3 to 4 weeks postinfection (p.i.) due to overwhelming virus replication in neurons. beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice had increased survival and decreased gray matter disease and virus replication compared to nontransgenic littermate controls. Both beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice developed chronic virus persistence in glial cells of the white matter of the spinal cord, with greater numbers of virus antigen-positive cells in beta(2)m(o) Abeta(o) DQ8 than in beta(2)m(o) Abeta(o) DQ6 mice. At day 45 p.i., the demyelinating lesions in the spinal cord of beta(2)m(o) Abeta(o) DQ8 were larger than those in the beta(2)m(o) Abeta(o) DQ6 mice. Earlier and more profound neurologic deficits were observed in beta(2)m(o) Abeta (o) DQ8 mice compared to beta(2)m(o) Abeta(o) DQ6 mice, although by 120 days p.i. both strains of mice showed similar extent of demyelination and neurologic deficits. Delayed-type hypersensitivity and antibody responses to TMEV demonstrated that the mice mounted class II-mediated cellular and humoral immune responses. The results are consistent with the hypothesis that rates of progression of demyelination and neurologic deficits are related to the differential ability of DQ6 and DQ8 transgenes to modulate the immune response and control virus.
Collapse
Affiliation(s)
- K D Pavelko
- Department of Immunology, Mayo Clinic and Mayo Foundation, Rochester, Minnesota, 55905, USA
| | | | | | | | | |
Collapse
|
37
|
Drescher KM, Murray PD, Lin X, Carlino JA, Rodriguez M. TGF-beta 2 reduces demyelination, virus antigen expression, and macrophage recruitment in a viral model of multiple sclerosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3207-13. [PMID: 10706712 DOI: 10.4049/jimmunol.164.6.3207] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGF-beta 2 is a potent immunoregulatory mediator that influences B cell, T cell, and macrophage function. To test whether this cytokine alters pathology in a model of virus-induced demyelinating disease, we treated SJL/J mice with TGF-beta 2 either before or after infection with Theiler's murine encephalomyelitis virus. Treatment continued three times weekly through day 35 postinfection. TGF-beta 2 administration resulted in significantly smaller lesions and fewer virus Ag-positive cells in the spinal cords of infected SJL/J mice. Mice treated with TGF-beta 2 had similar levels of virus-specific IgG as infected, control-treated mice. TGF-beta 2 administration significantly increased the level of non-virus-specific activated CTLs, but had no effect on virus-specific CTLs. TUNEL revealed a decrease in the number of apoptotic nuclei in the spinal cord white matter of mice treated in vivo with TGF-beta 2. Immunostaining with an Ab to F4/80 revealed that TGF-beta 2-treated mice had significantly fewer F4/80-positive cells in the white matter of the spinal cord as compared with infected control-treated mice. These data suggest that TGF-beta 2 may control virus-induced demyelination via an immunomodulatory mechanism that reduces macrophage infiltration.
Collapse
Affiliation(s)
- K M Drescher
- Departments of Neurology and Immunology, Mayo Clinic/Foundation, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
38
|
Drescher KM, Johnston SL, Hogancamp W, Nabozny GH, David CS, Rimm IJ, Wettstein PJ, Rodriguez M. V(beta)8(+) T cells protect from demyelinating disease in a viral model of multiple sclerosis. Int Immunol 2000; 12:271-80. [PMID: 10700462 DOI: 10.1093/intimm/12.3.271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Previous studies illustrated the influence of T cell subsets on susceptibility or resistance to demyelination in the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. Genetic segregation analysis showed a correlation with disease phenotype in this model with particular V(beta) genes. In this study we investigated the contribution of specific V(beta) TCR to the pathogenesis of virus-induced demyelinating disease. Spectratype analysis of cells infiltrating the CNS early in infection demonstrated an over-representation of V(beta)8(+) T cells in mice expressing a susceptible H-2 haplotype. We infected transgenic mice expressing the V(beta)8.2 TCR directed against a non-TMEV antigen and found an increase in demyelinating disease in mice of either susceptible or resistant background compared with littermate controls. In addition, depletion studies with an anti-V(beta)8-specific antibody in both susceptible (B10.Q) and resistant (C57BL/6) mice resulted in increased demyelination. TCR analysis of VP2-specific cytotoxic T cell clones from mice with a resistant genotype identified only the V(beta)8.1 TCR, suggesting that limited T cell diversity is critical to TMEV clearance. Together, these results support a protective role for V(beta)8(+) T cells in virus-induced demyelinating disease.
Collapse
Affiliation(s)
- K M Drescher
- Departments of Neurology and Immunology, Mayo Clinic/Foundation, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Palma JP, Yauch RL, Lang S, Kim BS. Potential Role of CD4+ T Cell-Mediated Apoptosis of Activated Astrocytes in Theiler’s Virus-Induced Demyelination. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.11.6543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Intracerebral inoculation of Theiler’s murine encephalomyelitis virus (TMEV) into susceptible mouse strains results in a chronic, immune-mediated demyelinating disease similar to human multiple sclerosis. Here, we examined the role of astrocytes as an APC population in TMEV-induced demyelination and assessed the potential consequences of T cell activation following Ag presentation. IFN-γ-pretreated astrocytes were able to process and present all the predominant T cell epitopes of TMEV to virus-specific T cell hybridomas, clones, as well as bulk T cells. Despite low levels of proliferation of T cells due to prostaglandins produced by astrocytes, such Ag presentation by activated astrocytes induced the production of IFN-γ, a representative proinflammatory cytokine, in TMEV-specific Th cell clones derived from the CNS of virus-infected mice. Furthermore, these Th cell clones mediate lysis of the astrocytes in vitro in a Fas-dependent mechanism. TUNEL staining of CNS tissue demonstrates the presence of apoptotic GFAP+ cells in the white matter of TMEV-infected mice. These results strongly suggest that astrocytes could play an important role in the pathogenesis of TMEV-induced demyelination by activating T cells, subsequently leading to T cell-mediated apoptosis of astrocytes and thereby compromising the blood-brain barrier.
Collapse
Affiliation(s)
- JoAnn P. Palma
- Departments of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611
| | - Robert L. Yauch
- Departments of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611
| | - Sharon Lang
- Departments of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611
| | - Byung S. Kim
- Departments of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611
| |
Collapse
|
40
|
McCright IJ, Tsunoda I, Whitby FG, Fujinami RS. Theiler's viruses with mutations in loop I of VP1 lead to altered tropism and pathogenesis. J Virol 1999; 73:2814-24. [PMID: 10074129 PMCID: PMC104039 DOI: 10.1128/jvi.73.4.2814-2824.1999] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Theiler's murine encephalomyelitis viruses are picornaviruses that can infect the central nervous system. The DA strain produces an acute polioencephalomyelitis followed by a chronic demyelinating disease in its natural host, the mouse. The ability of DA virus to induce a demyelinating disease renders this virus infection a model for human demyelinating diseases such as multiple sclerosis. Here we describe the generation and characterization of DA virus mutants that contain specific mutations in the viral capsid protein VP1 at sites believed to be important contact regions for the cellular receptor(s). A mutant virus with a threonine-to-aspartate (T81D) substitution in VP1 loop I adjacent to the putative virus receptor binding site exhibited a large-plaque phenotype but had a slower replication cycle in vitro. When this mutant virus was injected into susceptible mice, an altered tropism was seen during the acute stage of the disease and the chronic demyelinating disease was not produced. A virus with a threonine-to-valine substitution (T81V) did not cause any changes in the pattern or extent of disease seen in mice, whereas a virus with a tryptophan substitution at this position (T81W) produced a similar acute disease but was attenuated for the development of the chronic disease. A change in amino acids in a hydrophobic patch located in the wall of the pit, VP1 position 91, to a hydrophilic threonine (V91T) resulted in a profound attenuation of the acute and chronic disease without persistence of virus. This report illustrates the importance of the loop I of VP1 and a site in the wall of the pit in pathogenesis and that amino acid substitutions at these sites result in altered virus-host interactions.
Collapse
Affiliation(s)
- I J McCright
- Departments of Neurology, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | |
Collapse
|
41
|
Renno T, Taupin V, Bourbonnière L, Verge G, Tran E, De Simone R, Krakowski M, Rodriguez M, Peterson A, Owens T. Interferon-gamma in progression to chronic demyelination and neurological deficit following acute EAE. Mol Cell Neurosci 1998; 12:376-89. [PMID: 9888990 DOI: 10.1006/mcne.1998.0725] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The cytokine interferon-gamma (IFNgamma) is implicated in the induction of acute CNS inflammation, but it is less clear what role if any IFNgamma plays in progression to chronic demyelination and neurological deficit. To address this issue, we have expressed IFNgamma in myelinating oligodendrocytes of transgenic mice. MHC I immunostaining and iNOS mRNA were upregulated in their CNS, but such transgenic mice showed no spontaneous CNS inflammation or demyelination, and the incidence, severity, and histopathology of experimental autoimmune encephalomyelitis (EAE) were similar to nontransgenic controls. In contrast to control mice, which remit from EAE with resolution of glial reactivity and leukocytic infiltration, transgenics showed chronic neurological deficits. While activated microglia/macrophages persisted in demyelinating lesions for over 100 days, CD4(+) T lymphocytes were no longer present in CNS. IFNgamma therefore may play a role in chronic demyelination and long-term disability following the induction of demyelinating disease. Because IFNgamma may have neural as well as immune-infiltrating origins, these findings generate a new perspective on its role in the CNS.
Collapse
Affiliation(s)
- T Renno
- Neuroimmunology Unit, Montreal Neurological Institute, 3801 University, Montreal, Quebec, H3A 2B4, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Inoue A, Koh CS, Yamazaki M, Yahikozawa H, Ichikawa M, Yagita H, Kim BS. Suppressive Effect on Theiler’s Murine Encephalomyelitis Virus-Induced Demyelinating Disease by the Administration of Anti-IL-12 Antibody. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.10.5586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
We examined the role of IL-12, a cytokine critical to the evolution of cellular responses, in the development of Theiler’s murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). Treatment with mAbs to IL-12, especially during the effector phase, resulted in significant suppression of the development of this disease both clinically and histologically. In mice treated with these mAbs, the production of inflammatory and Th1-derived cytokines such as TNF-α and IFN-γ in the spleen cells was decreased, and that of Th2-derived cytokines such as IL-4 and IL-10 was increased. The delayed type hypersensitivity and T cell proliferative response specific for TMEV were decreased by this treatment. These data suggest that IL-12 is critically involved in the pathogenesis of TMEV-IDD and that Abs to IL-12 could be a novel therapeutic approach in the clinical treatment of demyelinating diseases such as human multiple sclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Motoki Ichikawa
- †Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideo Yagita
- ‡Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan; and
| | - Byung S. Kim
- §Departments of Microbiology-Immunology and Pathology, Northwestern University Medical School, Chicago, IL 60611
| |
Collapse
|
43
|
Murray PD, Pavelko KD, Leibowitz J, Lin X, Rodriguez M. CD4(+) and CD8(+) T cells make discrete contributions to demyelination and neurologic disease in a viral model of multiple sclerosis. J Virol 1998; 72:7320-9. [PMID: 9696828 PMCID: PMC109956 DOI: 10.1128/jvi.72.9.7320-7329.1998] [Citation(s) in RCA: 101] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/1998] [Accepted: 06/05/1998] [Indexed: 11/20/2022] Open
Abstract
Following intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV), susceptible strains of mice (SJL and PLJ) develop virus persistence and demyelination similar to that found in human multiple sclerosis. Resistant strains of mice (C57BL/6) clear virus and do not develop demyelination. To resolve the controversy about the role of CD4(+) and CD8(+) T cells in the development of demyelination and neurologic deficits in diseases of the central nervous system, we analyzed TMEV infection in CD4- and CD8-deficient B6, PLJ, and SJL mice. Genetic deletion of either CD4 or CD8 from resistant B6 mice resulted in viral persistence and demyelination during the chronic stage of disease. Viral persistence and demyelination were detected in all strains of susceptible background. Although genetic deletion of CD8 had no effect on the extent of demyelination in susceptible strains, deletion of CD4 dramatically increased the degree of demyelination observed. Whereas strains with deletions of CD4 showed severe neurologic deficits, mice with deletions of CD8 showed minimal or no deficits despite demyelination. In all strains, deletion of CD4 but not CD8 resulted in a decreased delayed-type hypersensitivity response to viral antigen. We conclude that each T-cell subset makes a discrete and nonredundant contribution to protection from viral persistence and demyelination in resistant strains. In contrast, in susceptible strains, CD8(+) T cells do not provide protection against chronic demyelinating disease. Furthermore, in persistent TMEV infection of the central nervous system, neurologic deficits appear to result either from the absence of a protective class II-restricted immune response or from the presence of a pathogenic class I-restricted response.
Collapse
Affiliation(s)
- P D Murray
- Departments of Immunology, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
44
|
Drescher KM, Nguyen LT, Taneja V, Coenen MJ, Leibowitz JL, Strauss G, Hammerling GJ, David CS, Rodriguez M. Expression of the human histocompatibility leukocyte antigen DR3 transgene reduces the severity of demyelination in a murine model of multiple sclerosis. J Clin Invest 1998; 101:1765-74. [PMID: 9541508 PMCID: PMC508759 DOI: 10.1172/jci167] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The role of various MHC genes in determining the progression of multiple sclerosis (MS) remains controversial. The HLA-DR3 gene has been associated with benign relapsing MS in some genetic epidemiologic studies, but with disease progression in others. We induced demyelination in highly susceptible B10.M and B10.Q mice expressing the DR3 (HLA-DRB1*0301) transgene to determine directly the effects of a human transgene by infecting them with Theiler's murine encephalomyelitis virus (TMEV). DR3+ mice experienced a dramatic reduction in the extent and severity of demyelination compared with DR3- littermate controls, whereas anti-TMEV antibody titers, delayed-type hypersensitivity responses, and levels of infectious virus, virus antigen, and virus RNA were similar in both groups. To address a possible mechanism of how the human transgene is reducing virus-induced demyelination, we analyzed cytokine expression in the lesions and also determined whether B10.M mice can respond to peptides derived from the DR3 molecule. Intense staining for IFN-gamma and IL-4, T helper (TH) 1 and TH2 cytokines, respectively, was found in the lesions of TMEV-infected DR3- mice but not in the DR3+ transgenic mice at day 21 after infection. DR3 peptides elicited strong proliferative responses in B10.M mice but not in B10.M (DR3+) mice. These experiments are the first to demonstrate that a human class II DR gene can alter the severity of demyelination in an animal model of MS without influencing viral load. These experiments are consistent with a mechanism by which DR3 reduces demyelination by altering the cytokine expression in the lesions, possibly by deleting T cells involved in virus-induced pathology.
Collapse
Affiliation(s)
- K M Drescher
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Borrow P, Welsh CJ, Tonks P, Dean D, Blakemore WF, Nash AA. Investigation of the role of delayed-type-hypersensitivity responses to myelin in the pathogenesis of Theiler's virus-induced demyelinating disease. Immunology 1998; 93:478-84. [PMID: 9659218 PMCID: PMC1364124 DOI: 10.1046/j.1365-2567.1998.00459.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The contribution of autoimmune responses to the pathogenesis of Theiler's virus-induced demyelinating disease was investigated. Delayed-type hypersensitivity responses to myelin were examined in both symptomatic and asymptomatic mice at different times post-infection, in order to determine whether autoreactivity correlates with the development of demyelination. The results indicate that although autoimmune responses probably do not play a major role in the initiation of demyelination at early times post-infection, autoreactivity to myelin antigens dose eventually develop in symptomatic animals, perhaps through the mechanism of epitope spreading. Autoimmunity to myelin components is therefore an additional factor that may contribute to lesion progression in chronically diseased animals.
Collapse
Affiliation(s)
- P Borrow
- Edward Jenner Institute for Vaccine Research, Newbury, Berkshire, UK
| | | | | | | | | | | |
Collapse
|
46
|
Bureau JF, Drescher KM, Pease LR, Vikoren T, Delcroix M, Zoecklein L, Brahic M, Rodriguez M. Chromosome 14 contains determinants that regulate susceptibility to Theiler's virus-induced demyelination in the mouse. Genetics 1998; 148:1941-9. [PMID: 9560407 PMCID: PMC1460094 DOI: 10.1093/genetics/148.4.1941] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Theiler's murine encephalomyelitis virus causes a chronic demyelinating disease in susceptible strains of mice that is similar to human multiple sclerosis. Several nonmajor histocompatibility complex-linked genes have been implicated as determinants of susceptibility or resistance to either demyelination or virus persistence. In this study, we used linkage analysis of major histocompatibility complex identical H-2d (DBA/2J x B10.D2) F2 intercross mice to identify loci associated with susceptibility to virus-induced demyelinating disease. In a 20-cM region on chromosome 14, we identified four markers, D14Mit54, D14Mit60, D14Mit61, and D14Mit90 that are significantly associated with demyelination. Because two peaks were identified, one near D14Mit54 and one near D14Mit90, it is possible that two loci in this region are involved in controlling demyelination.
Collapse
Affiliation(s)
- J F Bureau
- Unite des Virus Lents, Centre National de la Recherche Scientifique ERS 572, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin X, Sato S, Patick AK, Pease LR, Roos RP, Rodriguez M. Molecular characterization of a nondemyelinating variant of Daniel's strain of Theiler's virus isolated from a persistently infected glioma cell line. J Virol 1998; 72:1262-9. [PMID: 9445026 PMCID: PMC124604 DOI: 10.1128/jvi.72.2.1262-1269.1998] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/1997] [Accepted: 10/16/1997] [Indexed: 02/05/2023] Open
Abstract
Wild-type Daniel's strain of Theiler's virus (wt-DA) induces a chronic demyelination in susceptible mice which is similar to multiple sclerosis. A variant of wt-DA (designated DA-P12) generated during the 12th passage of persistent infection of a G26-20 glioma cell line failed to persist and induce demyelination in SJL/J mice. To identify the determinants responsible for this change in phenotype, we sequenced the capsid coding sequence (nucleotides [nt] 2991 to 3994) and found three mutations in VP1: residues 99 (Gly to Ser), 100 (Gly to Asp), and 103 (Asn to Lys). To study the role of these mutations in neurovirulence and demyelination, we prepared a recombinant virus, DAP-1C-2A/DA, with replacement of wt-DA nt 2991 to 3994 with the corresponding region of DA-P12, and viruses with individual point mutations at VP1 residues 99(Ser), 100(Asp), and 103(Lys). DAP-1C-2A/DA and viruses with a mutation at VP1 residue 99 or 100 (but not 103) completely attenuated the ability of wt-DA to induce demyelination. Failure to induce demyelination was not due to a general failure in growth, since DA-P12 and other mutant viruses lysed L-2 cells in vitro as effectively as wt-DA. The change in disease phenotype was independent of the specific B- or T-cell immune recognition because a decrease in the neurovirulence of mutant viruses was observed in neonatal mice and immune-deficient RAG1 -/- mice. This difference in neurovirulence is not the complete explanation for the failure of DA-P12 to demyelinate, since virus with a mutation at residue 103(Lys) had decreased neurovirulence but did induce demyelination. Therefore, point mutation at VP1 residue 99 or 100 altered the ability of wt-DA to demyelinate, perhaps related to a disruption in interaction between virus and receptor on certain neural cells.
Collapse
Affiliation(s)
- X Lin
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | |
Collapse
|
48
|
Kim BS, Yauch RL, Bahk YY, Kang JA, Dal Canto MC, Hall CK. A spontaneous low-pathogenic variant of Theiler's virus contains an amino acid substitution within the predominant VP1(233-250) T-cell epitope. J Virol 1998; 72:1020-7. [PMID: 9444995 PMCID: PMC124573 DOI: 10.1128/jvi.72.2.1020-1027.1998] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/1997] [Accepted: 10/30/1997] [Indexed: 02/05/2023] Open
Abstract
Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated demyelination after intracerebral inoculation of the virus into susceptible mouse strains. We isolated from a TMEV BeAn 8386 viral stock, a low-pathogenic variant which requires greater than a 10,000-fold increase in viral inoculation for the manifestation of detectable clinical signs. Intracerebral inoculation of this variant virus induced a strong, long-lasting, protective immunity from the demyelinating disease caused by pathogenic TMEV. The levels of antibodies to the whole virus as well as to the major linear epitopes were similar in mice infected with either the variant or wild-type virus. However, persistence of the variant virus in the central nervous system (CNS) of mice was significantly lower than that of the pathogenic virus. In addition, the T-cell response to the predominant VP1 (VP1(233-250)) epitope in mice infected with the variant virus was significantly weaker than that in mice infected with the parent virus, while similar T-cell responses were induced against another predominant epitope (VP2(74-86)). Further analyses indicated that a change of lysine to arginine at position 244 of VP1, which is the only amino acid difference in the P1 region, is responsible for such differential T-cell recognition. Thus, the difference in the T-cell reactivity to this VP1 region as well as the low level of viral persistence in the CNS may account for the low pathogenicity of this spontaneous variant virus.
Collapse
Affiliation(s)
- B S Kim
- Department of Microbiology-Immunology, and Institute for Neuroscience, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Drescher KM, Pease LR, Rodriguez M. Antiviral immune responses modulate the nature of central nervous system (CNS) disease in a murine model of multiple sclerosis. Immunol Rev 1997; 159:177-93. [PMID: 9416511 DOI: 10.1111/j.1600-065x.1997.tb01015.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The spectrum of disease is influenced by factors related to both the pathogen and the host, as well as the end points used in defining disease. In this article, the issue of disease resistance versus susceptibility will be examined in the framework that genetic manipulation of either the pathogen or the host immune response alters the balance from disease protection towards pathogenesis. The response of the host may trigger both a protective and a pathogenic immune response. The failure to mount a protective immune response predisposes the pathogen to persistence, which then becomes the target for immunopathology. This review will examine the factors involved both in virus-mediated pathogenesis and in disease protection in the Theiler's model of human multiple sclerosis. By manipulating the character of the virus pathogen and the specificity of the immune response, the entire spectrum of human demyelinating disease is reproduced.
Collapse
Affiliation(s)
- K M Drescher
- Department of Immunology, Mayo Clinic/Foundation, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
50
|
Inoue A, Koh CS, Yamazaki M, Yanagisawa N, Ishihara Y, Kim BS. Fibrin deposition in the central nervous system correlates with the degree of Theiler's murine encephalomyelitis virus-induced demyelinating disease. J Neuroimmunol 1997; 77:185-94. [PMID: 9258249 DOI: 10.1016/s0165-5728(97)00072-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We examined the role of coagulation-fibrinolysis system in Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). The degree of fibrin deposition around the vessels in the spinal cord was significantly higher in susceptible SJL/J mice on 30 days post intracerebral injection (i.c.) than resistant C57BL/6 mice on 30 days post i.c. or mock infected SJL/J mice. Treatment with batroxobin (30 BU/kg/day), which is a thrombin-like defibrinogenating enzyme, causing a profound degree of afibrinogenemia, suppressed clinical signs of TMEV-IDD. Plasma fibrinogen concentration was significantly decreased in batroxobin-treated mice. Histologically, though the degree of perivascular mononuclear cell infiltration in the spinal cord was not suppressed in batroxobin-treated mice compared to saline-treated control mice, fibrin deposition was markedly suppressed in batroxobin-treated mice. These findings suggest that batroxobin suppresses TMEV-IDD through its defibrination effect, and provide evidence that CNS-associated deposition of fibrin and ensuing fibrinolysis, together with increased permeability of the blood-brain barrier (BBB), are prerequisite events for clinical manifestations of TMEV-IDD.
Collapse
Affiliation(s)
- A Inoue
- Department of Medicine (Neurology), Shinshu University School of Medicine, Matsumoto, Japan
| | | | | | | | | | | |
Collapse
|