Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Adv Virus Res ; 56: 199-217, 2001.
Article in English | MEDLINE | ID: mdl-11450300

ABSTRACT

Epidemiological studies indicate that host immunogenetics and history of infection, particularly by viruses, may be a necessary cofactor for the induction of a variety of autoimmune diseases. To date, however, there is no clear-cut evidence, either in experimental animal models or in human autoimmune disease, that supports either molecular mimicry (Wucherpfennig and Strominger, 1995; Fujinami and Oldstone, 1985) or a role for superantigens (Scherer et al., 1993) in the initiation of T cell-mediated autoimmunity. In contrast, the current data provide compelling evidence in support of a major role for epitope spreading in the induction of myelin-specific autoimmunity in mice persistently infected with TMEV. It is significant that two picornaviruses closely related to TMEV, coxsackievirus (Rose and Hill, 1996) and encephalomyocarditis virus (EMCV) (Kyu et al., 1992), have been similarly shown to persist (either the viral RNA or the infectious virus) in their target organs and have been associated with the development of chronic autoimmune diseases, including myocarditis and diabetes. Thus, inflammatory responses induced by viruses that trigger proinflammatory Th1 responses, and have the ability to persist in genetically susceptible hosts, may lead to chronic organ-specific autoimmune disease via epitope spreading. Epitope spreading has important implications for the design of antigen-specific therapies for the potential treatment of MS and other autoimmune diseases. This process indicates that autoimmune diseases are evolving entities and that the specificity of the effector autoantigen-specific T cells varies during the chronic disease process. Our experiments employing tolerance in R-EAE clearly indicate that antigen-specific treatment of ongoing disease is possible for preventing disease relapses, provided the proper relapse-associated epitope is targeted (Vanderlugt et al., 1999). However, the ability to identify relapse-associated epitopes in humans will be a difficult task because immunodominance will vary in every individual. The use of costimulatory antagonists that can induce anergy without requiring prior knowledge of the exact epitopes (Miller et al., 1995b), or the use of therapies that induce bystander suppression (Nicholson et al., 1997; Brocke et al., 1996), may thus be more practical current alternative therapies for the treatment of human autoimmune disease.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases of the Nervous System/etiology , Autoimmune Diseases of the Nervous System/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Central Nervous System/immunology , Central Nervous System/virology , Myelin Sheath/immunology , Animals , Autoimmune Diseases of the Nervous System/immunology , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/pathology , Demyelinating Diseases/etiology , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Disease Models, Animal , Epitopes/immunology , Humans , Mice , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Theilovirus/pathogenicity , Theilovirus/physiology
2.
J Clin Invest ; 107(8): 995-1006, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11306603

ABSTRACT

Initial migration of encephalitogenic T cells to the central nervous system (CNS) in relapsing experimental autoimmune encephalomyelitis (R-EAE), an animal model of multiple sclerosis (MS), depends on the interaction of the alpha4 integrin (VLA-4) expressed on activated T cells with VCAM-1 expressed on activated cerebrovascular endothelial cells. Alternate homing mechanisms may be employed by infiltrating inflammatory cells after disease onset. We thus compared the ability of anti-VLA-4 to regulate proteolipid protein (PLP) 139-151-induced R-EAE when administered either before or after disease onset. Preclinical administration of anti-VLA-4 either to naive recipients of primed encephalitogenic T cells or to mice 1 week after peptide priming, i.e., before clinical disease onset, inhibited the onset and severity of clinical disease. In contrast, Ab treatment either at the peak of acute disease or during remission exacerbated disease relapses and increased the accumulation of CD4(+) T cells in the CNS. Most significantly, anti-VLA-4 treatment either before or during ongoing R-EAE enhanced Th1 responses to both the priming peptide and endogenous myelin epitopes released secondary to acute tissue damage. Collectively, these results suggest that treatment with anti-VLA-4 Ab has multiple effects on the immune system and may be problematic in treating established autoimmune diseases such as MS.


Subject(s)
Antibodies, Monoclonal/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrins/immunology , Receptors, Lymphocyte Homing/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cell Differentiation , Cell Division , Central Nervous System/immunology , Cytokines/genetics , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Immunotherapy/methods , Integrin alpha4beta1 , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger , Recurrence , T-Lymphocytes/immunology , Th1 Cells/cytology , Vascular Cell Adhesion Molecule-1/genetics
3.
J Immunol ; 165(9): 5304-14, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11046065

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease is a chronic-progressive, immune-mediated CNS demyelinating disease and a relevant model of multiple sclerosis. Myelin destruction is initiated by TMEV-specific CD4(+) T cells targeting persistently infected CNS-resident APCs leading to activation of myelin epitope-specific CD4(+) T cells via epitope spreading. We examined the temporal development of virus- and myelin-specific T cell responses and acquisition of virus and myelin epitopes by CNS-resident APCs during the chronic disease course. CD4(+) T cell responses to virus epitopes arise within 1 wk after infection and persist over a >300-day period. In contrast, myelin-specific T cell responses are first apparent approximately 50-60 days postinfection, appear in an ordered progression associated with their relative encephalitogenic dominance, and also persist. Consistent with disease initiation by virus-specific CD4(+) T cells, CNS mononuclear cells from TMEV-infected SJL mice endogenously process and present virus epitopes throughout the disease course, while myelin epitopes are presented only after initiation of myelin damage (>50-60 days postinfection). Activated F4/80(+) APCs expressing high levels of MHC class II and B7 costimulatory molecules and ingested myelin debris chronically accumulate in the CNS. These results suggest a process of autoimmune induction in which virus-specific T cell-mediated bystander myelin destruction leads to the recruitment and activation of infiltrating and CNS-resident APCs that process and present endogenous myelin epitopes to autoreactive T cells in a hierarchical order.


Subject(s)
Antigen Presentation , Antigen-Presenting Cells/immunology , Autoantigens/immunology , Epitopes, T-Lymphocyte/immunology , Myelin Proteolipid Protein/immunology , Spinal Cord/immunology , Th1 Cells/immunology , Theilovirus/immunology , Amino Acid Sequence , Animals , Antigen-Presenting Cells/metabolism , Autoantigens/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cardiovirus Infections/immunology , Cardiovirus Infections/pathology , Cell Count , Cells, Cultured , Chronic Disease , Disease Progression , Epitopes, T-Lymphocyte/metabolism , Female , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Inbred Strains , Molecular Sequence Data , Myelin Proteolipid Protein/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Time Factors
4.
J Neuroimmunol ; 109(2): 173-80, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10996219

ABSTRACT

During the progression of relapsing experimental autoimmune encephalomyelitis (R-EAE), in SJL mice, disease relapses are mediated by T cells specific for non-cross-reactive myelin epitopes, a process termed 'epitope spreading'. CTLA-4, a negative regulator of T cell function modulates R-EAE, in that CTLA-4 blockade exacerbates clinical R-EAE. Herein, we show that CTLA-4-mediated signaling negatively regulates the dynamic spread of autoreactive T cell responses during the course of autoimmune disease. Anti-CTLA-4 mAb, administration at various points during the progression of R-EAE exacerbated subsequent clinical disease and enhanced T cell reactivity to both inducing and relapse-associated epitopes. In addition, CTLA-4 blockade during acute disease inhibited clinical remission. Thus, CTLA-4-mediated events are critical for intrinsic regulation of epitope spreading during autoimmune disease.


Subject(s)
Antigens, Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Epitopes, T-Lymphocyte/immunology , Immunoconjugates , Abatacept , Acute Disease , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD , Autoantigens/immunology , CTLA-4 Antigen , Cross Reactions , Disease Models, Animal , Flow Cytometry , Hypersensitivity, Delayed/immunology , Immunization , Mice , Mice, Inbred Strains , Molecular Sequence Data , Multiple Sclerosis, Relapsing-Remitting/immunology , Myelin Basic Protein/chemistry , Myelin Basic Protein/immunology , Phospholipids/chemistry , Phospholipids/immunology , Recurrence , Remission, Spontaneous , T-Lymphocytes/immunology
5.
J Neuroimmunol ; 106(1-2): 43-52, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10814781

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) is a picornavirus which induces an immune-mediated demyelinating disease in susceptible strains of mice and serves as a relevant animal model for multiple sclerosis. Treatment with low dose irradiation prior to infection with the BeAn strain of TMEV renders the genetically resistant BALB/cByJ (C/cByJ) mice susceptible to disease. Previous studies have shown that disease resistance in the C/cByJ is mediated by a 'regulatory' CD8(+) T cell population, which does not appear to function via a cytolytic mechanism. We show here that TMEV-specific CD4(+) T cell blasts transferred into susceptible, irradiated C/cByJ accelerate clinical disease and enhance TMEV-specific DTH and proliferation in these animals. Significantly, CD8(+) cells from infected, resistant C/cByJ mice specifically downregulate the in vivo disease potentiation and diminish virus specific DTH, and proliferative and pro-inflammatory cytokine responses (IFNgamma and IL-2) in recipients of TMEV-specific CD4(+) T cell blasts. These results indicate that TMEV infection of resistant C/cByJ mice induces a radiosensitive population of regulatory CD8(+) T cells which actively downregulate inherent Th1 responses which have disease initiating potential.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cardiovirus Infections/etiology , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Theilovirus/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Division/physiology , Cytokines/metabolism , Disease Susceptibility , Down-Regulation , Hypersensitivity, Delayed/physiopathology , Hypersensitivity, Delayed/virology , Mice , Mice, Inbred BALB C , Th1 Cells/metabolism
6.
J Autoimmun ; 14(3): 205-11, 2000 May.
Article in English | MEDLINE | ID: mdl-10756082

ABSTRACT

PLP139-151-induced relapsing experimental autoimmune encephalomyelitis (R-EAE) in SJL mice is a Th1-mediated autoimmune demyelinating disease model for multiple sclerosis (MS) in which the primary disease relapse is mediated by T cells specific for the endogenous PLP178-191 epitope. This complex inflammatory process requires the co-ordinated expression of a wide variety of immune-related genes active at a variety of stages of the autoimmune process which are regulated, in part, by the transcription factor nuclear factor (NF)-kappaB which is activated via the ubiquitin-proteasome pathway. We asked if in vivo administration of a selective inhibitor of the ubiquitin-proteasome pathway, PS-519, which downregulates activation of NF-kappaB, could downregulate ongoing R-EAE. Administration of PS-519 during the remission phase, following acute clinical disease was effective in significantly reducing the incidence of clinical relapses, CNS histopathology, and T cell responses to both the initiating and relapse-associated PLP epitopes. The inhibition of clinical disease was dependent upon continuous administration of PS-519 in that recovery of T cell function and onset of disease relapses developed within 10-14 days of drug withdrawal. The data suggest that targeting the ubiquitin proteasome pathway, in particular NF-kappaB, may offer a novel and efficacious approach for the treatment of progressive autoimmune diseases, including MS.


Subject(s)
Cysteine Endopeptidases/immunology , Cysteine Proteinase Inhibitors/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multienzyme Complexes/immunology , Animals , Cysteine Proteinase Inhibitors/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Epitopes, T-Lymphocyte/immunology , Female , Mice , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Proteasome Endopeptidase Complex , Recurrence , Spinal Cord/immunology , T-Lymphocytes/immunology
7.
J Neuroimmunol ; 104(1): 79-84, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10683517

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) infection produces a chronic inflammatory disease of the spinal cord white matter, with striking similarities to both experimental allergic encephalomyelitis (EAE) and human multiple sclerosis (MS). The first phase of demyelination in this model appears to be dependent on a delayed-type hypersensitivity (DTH) response to viral antigens, driven by CD4+, Th1 lymphocytes. Macrophages, recruited in the infected CNS, would be responsible for most of the myelin damage. Recently, new populations of CD4+ lymphocytes were demonstrated in infected mice, this time with specificity for myelin antigens, particularly PLP. This suggests that, in the chronic phase of the disease, an autoimmune mechanism of demyelination, similar to EAE, may participate in the process of myelin destruction. The present study represents a first step in exploring the functional activity of these anti-myelin lymphocytes that emerge during the chronic phase of the disease. Lymphocytes were removed from chronically infected animals, they were stimulated with the major PLP encephalitogenic epitope for SJL/J mice, and they were added to organotypic myelinated spinal cord cultures for different lengths of time. Results show that lymphocytes stimulated with the major PLP epitope have a powerful capacity for demyelinating these cultures, while MBP stimulated lymphocytes and lymphocytes from control animals do not. This study, suggests that the anti-myelin response that emerges during the chronic phase of the infection is functionally active. A similar phenomenon of epitope spreading from virus to organ specific antigens may take place in humans and be involved in a number of immune-mediated diseases, including MS.


Subject(s)
Cardiovirus Infections/physiopathology , Demyelinating Diseases/physiopathology , Lymphocytes/physiology , Myelin Proteolipid Protein/pharmacology , Myelin Sheath/physiology , Theilovirus , Animals , Cardiovirus Infections/pathology , Cells, Cultured , Chronic Disease , Demyelinating Diseases/pathology , Encephalitis/immunology , Epitopes/pharmacology , Immunization , Mice , Mice, Inbred Strains , Myelin Basic Protein/pharmacology , Myelin Proteolipid Protein/immunology , Organ Culture Techniques , Ovalbumin/immunology
8.
J Immunol ; 164(2): 670-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10623809

ABSTRACT

Relapsing experimental autoimmune encephalomyelitis (R-EAE) is a CD4+ T cell-mediated demyelinating disease model for multiple sclerosis. Myelin destruction during the initial relapsing phase of R-EAE in SJL mice initiated by immunization with the proteolipid protein (PLP) epitope PLP139-151 is associated with activation of T cells specific for the endogenous, non-cross-reactive PLP178-191 epitope (intramolecular epitope spreading), while relapses in R-EAE induced with the myelin basic protein (MBP) epitope MBP84-104 are associated with PLP139-151-specific responses (intermolecular epitope spreading). Here, we demonstrate that T cells specific for endogenous myelin epitopes play the major pathologic role in mediating clinical relapses. T cells specific for relapse-associated epitopes can serially transfer disease to naive recipients and are demonstrable in the CNS of mice with chronic R-EAE. More importantly, induction of myelin-specific tolerance to relapse-associated epitopes, by i.v. injection of ethylene carbodiimide-fixed peptide-pulsed APCs, either before disease initiation or during remission from acute disease effectively blocks the expression of the initial disease relapse. Further, blockade of B7-1-mediated costimulation with anti-B7-1 F(ab) during disease remission from acute PLP139-151-induced disease prevents clinical relapses by inhibiting activation of PLP178-191-specific T cells. The protective effects of anti-B7-1 F(ab) treatment are long-lasting and highly effective even when administered following the initial relapsing episode wherein spreading to a MBP epitope (MBP84-104) is inhibited. Collectively, these data indicate that epitope spreading is B7-1 dependent, plays a major pathologic role in disease progression, and follows a hierarchical order associated with the relative encephalitogenic dominance of the myelin epitopes (PLP139-151 > PLP178-191 > MBP84-104).


Subject(s)
Autoantigens/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Epitopes, T-Lymphocyte/physiology , Adoptive Transfer , Amino Acid Sequence , Animals , Antibodies, Monoclonal/administration & dosage , Autoantigens/administration & dosage , Autoantigens/biosynthesis , B7-1 Antigen/immunology , B7-1 Antigen/physiology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/biosynthesis , Female , Immune Tolerance/immunology , Immunodominant Epitopes/physiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Myelin Basic Protein/administration & dosage , Myelin Basic Protein/immunology , Myelin Basic Protein/physiology , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/immunology , Myelin Proteolipid Protein/physiology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/physiology , Recurrence , T-Lymphocytes/transplantation , Time Factors
9.
J Clin Invest ; 104(5): 599-610, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10487774

ABSTRACT

The mechanisms underlying the initiation of virus-induced autoimmune disease are not well understood. Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), a mouse model of multiple sclerosis, is initiated by TMEV-specific CD4(+) T cells targeting virally infected central nervous system-resident (CNS-resident) antigen-presenting cells (APCs), leading to chronic activation of myelin epitope-specific CD4(+) T cells via epitope spreading. Here we show that F4/80(+), I-A(s+), CD45(+) macrophages/microglia isolated from the CNS of TMEV-infected SJL mice have the ability to endogenously process and present virus epitopes at both acute and chronic stages of the disease. Relevant to the initiation of virus-induced autoimmune disease, only CNS APCs isolated from TMEV-infected mice with preexisting myelin damage, not those isolated from naive mice or mice with acute disease, were able to endogenously present a variety of proteolipid protein epitopes to specific Th1 lines. These results offer a mechanism by which localized virus-induced, T cell-mediated inflammatory myelin destruction leads to the recruitment/activation of CNS-resident APCs that can process and present endogenous self epitopes to autoantigen-specific T cells, and thus provide a mechanistic basis by which epitope spreading occurs.


Subject(s)
Antigen Presentation , Autoimmune Diseases/immunology , Cardiovirus Infections/immunology , Central Nervous System/immunology , Demyelinating Diseases/immunology , Disease Models, Animal , Epitopes/immunology , Multiple Sclerosis/immunology , Myelin Proteolipid Protein/immunology , Theilovirus , Amino Acid Sequence , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/pathology , Autoimmune Diseases/virology , CD4-Positive T-Lymphocytes/immunology , Cardiovirus Infections/complications , Cells, Cultured , Central Nervous System/pathology , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/virology , Female , Macrophages/immunology , Mice , Mice, Inbred Strains , Microglia/immunology , Molecular Sequence Data , Multiple Sclerosis/virology , Peptide Fragments/immunology , Spinal Cord/pathology , Th1 Cells/immunology
10.
J Neuroimmunol ; 98(2): 136-46, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10430047

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) is an excellent model for human multiple sclerosis. Within the BALB/c strain, BALB/cAnNCr mice are susceptible while BALB/cByJ mice are resistant. BALB/cByJ mice become susceptible when irradiated. Adoptive transfer of CD8+ splenic T cells from resistant BALB/cByJ donors protect irradiated BALB/cByJ, as well as BALB/cAnNCr recipients, from development of TMEV-IDD. Anti-TMEV CTL activities in BALB/cAnNCr, BALB/cByJ and irradiated BALB/cByJ mice are comparable. A population of splenic CD4+ T cells in BALB/cByJ donors has also been identified which can protect both susceptible BALB/cAnNCr and irradiated BALB/cByJ recipients from TMEV-IDD via adoptive transfer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Demyelinating Diseases/immunology , Theilovirus/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , Cell Transplantation , Demyelinating Diseases/virology , Disease Susceptibility , Fibrosarcoma , Gamma Rays , Kinetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuroimmunomodulation/immunology , Spleen/cytology , Spleen/immunology , Spleen/radiation effects , Thymectomy , Tumor Cells, Cultured
11.
J Clin Invest ; 103(2): 281-90, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9916140

ABSTRACT

Relapsing experimental autoimmune encephalomyelitis (R-EAE) in the SJL mouse is a Th1-mediated autoimmune demyelinating disease model for human multiple sclerosis and is characterized by infiltration of the central nervous system (CNS) by Th1 cells and macrophages. Disease relapses are mediated by T cells specific for endogenous myelin epitopes released during acute disease, reflecting a critical role for epitope spreading in the perpetuation of chronic central CNS pathology. We asked whether blockade of the CD40-CD154 (CD40L) costimulatory pathway could suppress relapses in mice with established R-EAE. Anti-CD154 antibody treatment at either the peak of acute disease or during remission effectively blocked clinical disease progression and CNS inflammation. This treatment blocked Th1 differentiation and effector function rather than expansion of myelin-specific T cells. Although T-cell proliferation and production of interleukin (IL)-2, IL-4, IL-5, and IL-10 were normal, antibody treatment severely inhibited interferon-gamma production, myelin peptide-specific delayed-type hypersensitivity responses, and induction of encephalitogenic effector cells. Anti-CD154 antibody treatment also impaired the expression of clinical disease in adoptive recipients of encephalitogenic T cells, suggesting that CD40-CD154 interactions may be involved in directing the CNS migration of these cells and/or in their effector ability to activate CNS macrophages/microglia. Thus, blockade of CD154-CD40 interactions is a promising immunotherapeutic strategy for treatment of ongoing T cell-mediated autoimmune diseases.


Subject(s)
Antibodies/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunotherapy/methods , Membrane Glycoproteins/immunology , Multiple Sclerosis/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand , Cell Differentiation/immunology , Cell Division/immunology , Central Nervous System/immunology , Central Nervous System/pathology , Disease Models, Animal , Female , Hypersensitivity, Delayed/immunology , Inflammation/immunology , Interferon-gamma/immunology , Interleukins/immunology , Mice , Mice, Inbred Strains , Myelin Proteolipid Protein/immunology , Myelin Sheath/immunology , Peptide Fragments/immunology , Th1 Cells/immunology
12.
J Immunol ; 161(8): 4437-46, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9780223

ABSTRACT

Multiple sclerosis is an immune-mediated demyelinating disease of unknown etiology that presents with either a chronic-progressive or relapsing-remitting clinical course. Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) and relapsing-remitting experimental autoimmune encephalomyelitis (R-EAE) in the SJL/J mouse are both relevant murine CD4+ T cell-mediated demyelinating models that recapitulate the multiple sclerosis disease phenotypes. To determine the cellular and molecular basis for these observed differences in clinical course, we quantitatively analyzed the temporal expression of pro- and antiinflammatory cytokine mRNA expression in the central nervous system (CNS) and the phenotype of the inflammatory mononuclear infiltrates. TMEV-infected SJL/J mice expressed IFN-gamma, TNF-alpha, IL-10, and IL-4 mRNA during the preclinical phase, and their levels continued to increase throughout the duration of the chronic-progressive disease course. These data correlated with the continued presence of both CD4+ T cells and F4/80+ macrophages within the CNS infiltrates. In contrast, SJL/J mice with PLP(139-151)-induced R-EAE displayed a biphasic pattern of CNS expression for the proinflammatory cytokines, IFN-gamma and TNF-alpha, with expression peaking at the height of the acute phase and relapse(s). This pattern correlated with dynamic changes in the CD4+ T cell and F4/80+ macrophage populations during relapsing-remitting disease progression. Interestingly, IL-4 message was undetectable until disease remission(s), demonstrating its potential role in the intrinsic regulation of ongoing disease, whereas IL-10 was continuously expressed, arguing against a regulatory role in either disease. These data suggest that the kinetics of cytokine expression together with the nature of the persistent inflammatory infiltrates are major contributors to the differences in clinical course between TMEV-IDD and R-EAE.


Subject(s)
Central Nervous System/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Animals , Biomarkers , Central Nervous System/pathology , Cytokines/biosynthesis , Disease Models, Animal , Encephalomyelitis/immunology , Encephalomyelitis/pathology , Encephalomyelitis/virology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Multiple Sclerosis/pathology
13.
Immunol Rev ; 164: 63-72, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9795764

ABSTRACT

Epitope spreading is a process whereby epitopes distinct from and non-cross-reactive with an inducing epitope become major targets of an ongoing immune response. This phenomenon has been defined in experimental and natural situations as a consequence of acute or persistent infection and secondary to chronic tissue destruction that occurs during progressive autoimmune disease. We have investigated the functional significance of this process in the chronic stages of both autoimmune and virus-induced central nervous system (CNS) demyelinating disease models in the SJL/J mouse. During the relapsing-remitting course of experimental autoimmune encephalomyelitis (R-EAE) induced with defined encephalitogenic myelin peptides, CD4+ T cells specific for endogenous epitopes on both the initiating myelin protein (intramolecular epitope spreading) and distinct myelin proteins (intermolecular epitope spreading) are primed secondary to myelin destruction during acute disease and play a major functional role in mediating disease relapses. Similarly, epitope spreading to endogenous myelin epitopes appears to play a major functional role in the chronic-progressive course of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), a virus-induced CD4+ T-cell-mediated immunopathology. In TMEV-IDD, myelin destruction is initiated by virus-specific CD4+ T cells which target virus epitopes persisting in CNS-derived antigen-presenting cells. However, the chronic stage of this progressive disease is associated with the activation of CD4+ T cells specific for multiple myelin epitopes. In both models, the temporal course of T-cell activation occurs in a hierarchical order of epitope dominance, spreading first to the most immunodominant epitope and progressing to lesser immunodominant epitopes. In addition, epitope spreading in R-EAE is regulated predominantly by CD28/B7-1 co-stimulatory interactions, as antagonism of B7-1-mediated co-stimulation using anti-B7-1 F(ab) fragments is an effective ameliorative therapy for ongoing disease. The process of epitope spreading has obvious important implications for the design of antigen-specific therapies for the treatment of autoimmune disease since these therapies will have to identify and target endogenous self epitopes associated with chronic tissue destruction.


Subject(s)
Antigens, CD/immunology , Cardiovirus Infections/immunology , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunodominant Epitopes , Theilovirus , Animals , B7-1 Antigen/immunology , CD28 Antigens/immunology , Mice , Models, Immunological
14.
J Neuroimmunol ; 89(1-2): 10-8, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9726820

ABSTRACT

The B7/CD28:CTLA-4 costimulatory pathway plays a critical role in determining the fate of immune responses (activation vs. down-regulation) and is a highly promising therapeutic target for treating autoimmune diseases. In this review, we highlight the mechanisms by which this costimulatory pathway operates emphasizing the role of the different components in the pathogenesis of relapsing experimental autoimmune encephalomyelitis, a CD4 T cell-mediated autoimmune model of multiple sclerosis. The separate and distinct roles of B7-1, B7-2 and CTLA-4 in positive and negative regulation of autoimmune pathogenesis are considered and a working model is proposed.


Subject(s)
Antigens, Differentiation/immunology , B7-1 Antigen/immunology , CD28 Antigens/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoconjugates , Immunosuppressive Agents/immunology , Abatacept , Animals , Antigens, CD , CTLA-4 Antigen
15.
J Virol ; 72(10): 7762-71, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9733812

ABSTRACT

We examined the phenotype and function of cells infiltrating the central nervous system (CNS) of mice persistently infected with Theiler's murine encephalomyelitis virus (TMEV) for evidence that viral antigens are presented to T cells within the CNS. Expression of major histocompatibility complex (MHC) class II in the spinal cords of mice infected with TMEV was found predominantly on macrophages in demyelinating lesions. The distribution of I-As staining overlapped that of the macrophage marker sialoadhesin in frozen sections and coincided with that of another macrophage/microglial cell marker, F4/80, by flow cytometry. In contrast, astrocytes, identified by staining with glial fibrillary acidic protein, rarely expressed detectable MHC class II, although fibrillary gliosis associated with the CNS damage was clearly seen. The costimulatory molecules B7-1 and B7-2 were expressed on the surface of most MHC class II-positive cells in the CNS, at levels exceeding those found in the spleens of the infected mice. Immunohistochemistry revealed that B7-1 and B7-2 colocalized on large F4/80(+) macrophages/microglia in the spinal cord lesions. In contrast, CD4(+) T cells in the lesions expressed mainly B7-2, which was found primarily on blastoid CD4(+) T cells located toward the periphery of the lesions. Most interestingly, plastic-adherent cells freshly isolated from the spinal cords of TMEV-infected mice were able to process and present TMEV and horse myoglobin to antigen-specific T-cell lines. Furthermore, these cells were able to activate a TMEV epitope-specific T-cell line in the absence of added antigen, providing conclusive evidence for the endogenous processing and presentation of virus epitopes within the CNS of persistently infected SJL/J mice.


Subject(s)
Antigen-Presenting Cells/immunology , Central Nervous System/pathology , Macrophages/immunology , Theilovirus/isolation & purification , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Antigens, Viral/isolation & purification , Astrocytes/immunology , B7-1 Antigen/immunology , Central Nervous System/virology , Female , Flow Cytometry , Immunohistochemistry , Lymphocyte Activation , Mice , Th1 Cells/immunology , Theilovirus/immunology
16.
J Immunol ; 161(1): 192-9, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9647224

ABSTRACT

B7/CD28-mediated costimulation is a promising target for therapeutic intervention in autoimmune diseases. However, studies addressing the differential functional roles of B7-1 and B7-2 in several autoimmune models have resulted in conflicting data, perhaps due to the temporal dynamics of B7-1 and B7-2 surface expression on different cell types and/or at different sites during an autoimmune response. We examined the temporal expression of B7 costimulatory molecules in the CNS and in various lymphoid organs during the course of murine relapsing-remitting experimental autoimmune encephalomyelitis (R-EAE). Following immunization of SJL mice with the immunodominant proteolipid protein epitope, PLP139-151, surface expression of B7-1 was up-regulated on B cells, T cells, and macrophages, relative to B7-2, on CNS-infiltrating cells and on splenocytes. Similar enhancement in splenic B7-1 expression could be induced in SJL mice by the adoptive transfer of PLP139-151-specific cells or by immunization with CFA alone. These changes were not observed on lymph node cells, including those isolated from lymph nodes draining the immunization site, which maintained the predominant B7-2 expression pattern seen in naive mice. These phenotypic expression patterns correlated with the functional predominance of B7-1 in costimulating T cell activation when employing APCs from the spleen or CNS of mice with ongoing R-EAE, while B7-2 remained functionally predominant on lymph node APCs. Variation of phenotypic expression and functional dominance of costimulatory molecule expression in different lymphoid compartments during an active inflammatory autoimmune response has important implications in immune regulation, autoimmune pathogenesis, and therapeutic strategies.


Subject(s)
B7-1 Antigen/biosynthesis , B7-1 Antigen/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymph Nodes/immunology , Spleen/immunology , Up-Regulation/immunology , Animals , Antigens, CD/biosynthesis , Antigens, CD/physiology , B7-2 Antigen , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Lymph Nodes/cytology , Lymph Nodes/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred Strains , Organ Specificity/immunology , Recurrence , Spleen/cytology , Spleen/metabolism
17.
Autoimmunity ; 27(1): 13-28, 1998.
Article in English | MEDLINE | ID: mdl-9482204

ABSTRACT

Our previous work has shown that specific peripheral immune tolerance induced by the intravenous administration of ECDI-fixed, antigen-coupled syngeneic splenocytes is an extremely efficient method for prevention and treatment of chronic relapsing experimental autoimmune encephalomyelitis (R-EAE) in susceptible SJL/J mice. The current study examined the mechanisms by which unresponsiveness is induced in primed encephalitogenic T cells. The results indicate that the inhibition of MBP-specific T cells by the i.v. injection of MBP-coupled splenocytes is not due to the induction of antigen-specific regulatory T cells, but rather to the induction of anergy/deletion of the effector cells. This conclusion is supported by the findings that spleen or lymph node cells isolated from MBP-tolerant mice fail to inhibit the adoptive transfer of R-EAE in cotransfer assays, and that tolerance is not inhibited by prior thymectomy or prior treatment with cyclophosphamide or anti-CD8 monoclonal antibody. In contrast, we demonstrate that splenocytes from MBP-tolerized, asymptomatic mice have a significantly reduced ability to serially transfer R-EAE to naive secondary recipients following antigen re-activation in vitro, in the first several weeks following tolerization, but that the ability to serially transfer R-EAE returns to sham tolerant control levels within 1-2 months. We also demonstrate a significantly reduced precursor frequency of MBP-specific, IL-2-producing T cells in the MBP-tolerant within three days of treatment. Collectively, the data most closely support a model wherein inhibition of MBP-specific encephalitogenic CD4+ effector T cells by i.v. injected MBP-coupled splenocytes is due to the direct induction of anergy/deletion from which they can recover over time.


Subject(s)
Autoantigens/immunology , Clonal Anergy/immunology , Clonal Deletion/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin Basic Protein/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Autoantigens/administration & dosage , Cattle , Disease Models, Animal , Female , Mice , Mice, Inbred Strains , Myelin Basic Protein/administration & dosage , Rats , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/drug effects , Time Factors
18.
J Invest Dermatol ; 110(2): 103-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9457902

ABSTRACT

Autoimmune diseases are initiated when patients develop aberrant T and/or B cell responses against self proteins. These responses presumably are directed to single immunogenic epitopes on these proteins. Recent data in animal models of autoimmune diseases suggest that the targets of immune responses in autoimmunity do not remain fixed, but can be extended to include other epitopes on the same protein or other proteins in the same tissue, a phenomenon termed "epitope spreading." The "epitope spreading" phenomenon also applies to situations in which tissue damage from a primary inflammatory process causes the release and exposure of a previously "sequestered" antigen, leading to a secondary autoimmune response against the newly released antigen. In experimental autoimmune animal diseases, "epitope spreading" seems to have significant physiologic importance in determining the course and duration of disease. In this paper, we review the current concepts in animal models of autoimmune diseases in order to define the "epitope spreading" phenomenon, and we then propose how this phenomenon might play a significant role in the development and the course of autoimmune skin diseases. Hopefully, an understanding of "epitope spreading" will help the dermatology community to better understand the pathogenesis of autoimmune skin diseases and to rationally fashion disease-specific immune therapy in the future.


Subject(s)
Autoimmune Diseases/immunology , Epitopes/physiology , Skin Diseases/immunology , Animals , Disease Models, Animal , Humans
19.
J Neuroimmunol ; 79(2): 113-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9394783

ABSTRACT

PLP139-151-induced experimental autoimmune encephalomyelitis in the SJL mouse is a Th1-mediated inflammatory demyelinating disease characterized by a relapsing-remitting clinical course (R-EAE). Clinical relapses are mediated by T cells specific for a non-cross reactive secondary PLP epitope (PLP178-191) induced by epitope spreading. We have previously shown that B7-1 expression is upregulated in SJL mice undergoing R-EAE and in vivo treatment during remission with F(ab) fragments of anti-B7-1 mAb, blocked epitope spreading and disease progression. In contrast, the present study shows that treatment with intact anti-B7-1 mAb exacerbated clinical disease relapses and enhanced CNS demyelination. Anti-B7-1-treated mice showed enhanced in vivo delayed-type hypersensitivity (DTH) to the relapse-associated PLP178-191 epitope and responses to the immunodominant MBP84-104 epitope which are absent in the controls. Thus, ligation of B7-1 by intact mAbs has effects opposite to those of anti-B7-1 F(ab) fragments suggesting that the mAb is directly signaling through B7-1 expressed on T cells and/or APCs.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-1 Antigen/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Epitopes/drug effects , Animals , Female , Immunoglobulin Fab Fragments/pharmacology , Mice , Mice, Inbred Strains , Recurrence , Remission Induction
20.
Nat Med ; 3(10): 1133-6, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9334726

ABSTRACT

Multiple sclerosis (MS) is a T cell-mediated autoimmune demyelinating disease, which may be initiated by a virus infection. Theiler's murine encephalomyelitis virus (TMEV), a natural mouse pathogen, is a picornavirus that induces a chronic, CD4+ T cell-mediated demyelinating disease with a clinical course and histopathology similar to that of chronic progressive MS (ref. 3). Demyelination in TMEV-infected mice is initiated by a mononuclear inflammatory response mediated by virus-specific CD4+ T cells targeting virus, which chronically persists in the CNS (ref. 4-6). We show that beginning 3-4 weeks after disease onset, T-cell responses to multiple myelin autoepitopes arise in an ordered progression and may play a pathologic role in chronic disease. Kinetic and functional studies show that T-cell responses to the immunodominant myelin proteolipid protein epitope (PLP139-151) did not arise because of cross-reactivity between TMEV and self epitopes (that is, molecular mimicry), but because of de novo priming of self-reactive T cells to sequestered autoantigens released secondary to virus-specific T cell-mediated demyelination (that is, epitope spreading). Epitope spreading is an important alternate mechanism to explain the etiology of virus-induced organ-specific autoimmune diseases.


Subject(s)
Autoimmunity , Demyelinating Diseases/immunology , Epitopes/immunology , Myelin Proteins/immunology , Myelin Proteolipid Protein/immunology , Poliomyelitis/immunology , T-Lymphocytes/immunology , Theilovirus , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , Cross Reactions , Female , Inflammation , Kinetics , Mice , Mice, Inbred Strains , Molecular Sequence Data , Myelin Proteins/chemistry , Myelin Proteolipid Protein/chemistry , Poliomyelitis/physiopathology , Self Tolerance
SELECTION OF CITATIONS
SEARCH DETAIL
...