Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Virology ; 287(2): 301-9, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11531408

ABSTRACT

Both human and murine forms of nectin-1 (HveC, Prr1) can serve as entry receptors for several neurotropic herpesviruses, including herpes simplex viruses 1 and 2 (HSV-1, HSV-2), porcine pseudorabies virus (PRV), and bovine herpesvirus 1. HSV-1, HSV-2, and PRV can cause lethal neurological disease in mice whether inoculation is directly into the central nervous system or by peripheral routes. Expression of nectin-1 transcripts in cells of the adult mouse nervous system was assessed by in situ hybridization. Specific hybridization signals were detected in neurons in sensory, sympathetic, and parasympathetic ganglia of the peripheral nervous system. In addition, specific signals were observed in neurons of the ventral and dorsal horns of the spinal cord and of the brain stem, cerebellum, cerebral cortex, hippocampus, dentate gyrus, and olfactory bulb. These results show that the nectin-1 gene is widely transcribed in neurons in adult mouse. Nectin-1 is the only known receptor capable of mediating the entry of all three viruses, HSV-1, HSV-2, and PRV. Its pattern of expression in the nervous system suggests a key role in neurological disease caused by these viruses.


Subject(s)
Cell Adhesion Molecules/genetics , Neurons/metabolism , Animals , Cell Adhesion Molecules/metabolism , Central Nervous System/metabolism , Female , Ganglia/metabolism , Gene Expression , Herpesviridae/physiology , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Nectins , RNA/metabolism , Spinal Cord/metabolism , Transcription, Genetic
2.
J Neuroimmunol ; 118(2): 256-67, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11498260

ABSTRACT

The BeAn strain of Theiler's murine encephalomyelitis virus (TMEV) persists in the CNS and produces a chronic inflammatory demyelinating disease that is an animal model for human multiple sclerosis (MS). The mechanisms leading to TMEV-induced demyelination are still under study but most likely involve both immune-mediated and virus induced damage to cells in the CNS, both depending on viral persistence. It is therefore important to identify the cells in which continued virus production is permitted. In this study, we looked at virus infection in primary astrocytes, microglia and oligodendrocytes, derived from brains of neonatal susceptible SJL/J mice. As evidenced by Western blots and immunocytochemistry, we were able to detect viral antigens in all these brain-derived cells. In addition, we extended the study to spinal cord tissues from mice suffering TMEV-induced disease. Immunohistochemistry staining with anti-TMEV sera and antibodies to specific cell markers detected viral antigens in all these cells. We then asked the question whether viral antigen present in these cells, particularly in microglia/macrophages, represented true viral replication or not. By using different techniques, including immunoprecipitation experiments and the very sensitive method of negative RNA detection through RNase protection assay, we show that both astrocytes and oligodendroglia permit de novo viral replication and viral protein synthesis but with only minimal cytopathic effects. Of these two cell types, astrocytes carry the brunt of viral replication. In microglia, on the other hand, viral replication is restricted since only minimal amounts of negative RNA copies can be demonstrated, while there are clear signs that some of these cells undergo apoptosis. These findings show that the main cell for viral replication is the astrocyte, rather than the microglia/macrophage. Most of the viral antigen present in macrophages, therefore, is probably the result of phagocytosis, rather than actual viral replication. In view of the demonstrated presence of viral replication in astrocytes and of great amounts of viral antigens in microglia/macrophages, it is possible that both types of cells act as antigen presenting cells during this immunopathological disease.


Subject(s)
Astrocytes/virology , Cardiovirus Infections/virology , Demyelinating Diseases/virology , Microglia/pathology , Theilovirus/physiology , Virus Latency , Animals , Astrocytes/chemistry , Astrocytes/drug effects , Astrocytes/pathology , Blotting, Western , Cardiovirus Infections/pathology , Cells, Cultured , Central Nervous System Viral Diseases/pathology , Central Nervous System Viral Diseases/virology , Cricetinae , Cytokines/pharmacology , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Immunohistochemistry , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred Strains , Microglia/chemistry , Microglia/drug effects , Microglia/virology , Oligodendroglia/chemistry , Oligodendroglia/pathology , Oligodendroglia/virology , Precipitin Tests , RNA, Viral/analysis , Theilovirus/growth & development , Theilovirus/isolation & purification , Viral Proteins/analysis , Viral Proteins/biosynthesis , Virus Replication/drug effects
3.
J Clin Invest ; 108(2): 311-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11457884

ABSTRACT

Molecular mimicry is the process by which virus infection activates T cells that are cross-reactive with self antigens. Infection of SJL/J mice with the neurotropic picornavirus Theiler's murine encephalomyelitis virus (TMEV) leads to a progressive CD4(+) T cell-mediated demyelinating disease similar to multiple sclerosis. To study the potential of virus-induced molecular mimicry to initiate autoimmune demyelination, a nonpathogenic TMEV variant was engineered to encode a 30-mer peptide encompassing the immunodominant encephalitogenic myelin proteolipid protein (PLP139-151) epitope. Infection with the PLP139-151-encoding TMEV led within 10-14 days to a rapid-onset paralytic demyelinating disease characterized by PLP139-151-specific CD4(+) Th1 responses; insertion of a non-self ovalbumin sequence led to restoration of the normal late-onset disease. Early-onset disease was also observed in mice infected with a TMEV encoding PLP139-151 with an amino acid substitution at the secondary T cell receptor (TCR) contact residue (H147A), but not in mice infected with TMEV encoding a PLP139-151 substitution at the primary TCR contact (W144A). Most significantly, mice infected with TMEV encoding a Haemophilus influenzae mimic peptide, sharing only 6 of 13 amino acids with PLP139-151, displayed rapid-onset disease and developed cross-reactive PLP139-151-specific CD4(+) Th1 responses. To our knowledge, this is the first study showing that a naturally infectious virus encoding a myelin epitope mimic can directly initiate organ-specific T cell-mediated autoimmunity.


Subject(s)
Cardiovirus Infections/virology , Encephalitis, Viral/virology , Molecular Mimicry , Multiple Sclerosis/etiology , Myelin Proteolipid Protein/biosynthesis , Peptide Fragments/biosynthesis , Theilovirus/metabolism , Amino Acid Sequence , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , Cardiovirus Infections/immunology , Cross Reactions , Cytokines/analysis , Demyelinating Autoimmune Diseases, CNS/immunology , Disease Models, Animal , Encephalitis, Viral/immunology , Epitopes/chemistry , Mice , Molecular Sequence Data , Myelin Proteolipid Protein/chemistry , Myelin Proteolipid Protein/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Recombination, Genetic , Th1 Cells/immunology , Theilovirus/genetics
4.
Hum Pathol ; 31(10): 1274-98, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11070121

ABSTRACT

Acquired immune deficiency syndrome (AIDS), caused by human immunodeficiency virus type 1 (HIV-1), has claimed more than 10 million lives over the past 15 years. There are approximately 30 million HIV-positive people worldwide, 89% of whom reside in sub-Saharan Africa and Asia. The intricate relationship between the virus and HIV-related human multisystem pathology prompted scientists to modify many previously established concepts about infectious diseases and immunology, and to test new ones. The results of this work helped resolve many, albeit not all, long-standing problems concerning HIV-1 immune escape, its cellular tropism, and pathogenesis of HIV-related immunosuppression and nervous system disease. The most impressive advances have been made in antiretroviral drug treatment of HIV infection, which has resulted in dramatically reducing AIDS-related mortality, morbidity, and perinatal transmission. However, considering the magnitude of the worldwide HIV-AIDS pandemic, prohibitive cost and unusually exacting nature of combination drug treatment, as well as the emergence of drug-resistant HIV mutants, the disease and virus remain formidable and unpredictable, particularly in the area of prevention and vaccine development. Here, we have reviewed the most pertinent recently published studies of various aspects of HIV/AIDS intended to answer the following questions: what have we learned and what remains to be determined regarding this unorthodox viral disorder?


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Antigenic Modulation , HIV-1/immunology , Immune Tolerance , Nervous System Diseases/etiology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/virology , Acquired Immunodeficiency Syndrome/immunology , HIV-1/genetics , Humans , Lymphoid Tissue/virology , Muscle, Skeletal/virology , Mutagenesis , Nervous System/virology , Nervous System Diseases/immunology , Receptors, Chemokine/genetics , Viral Load
5.
J Virol ; 74(24): 11773-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090177

ABSTRACT

A cDNA encoding the murine homolog of human nectin-1alpha (also known as poliovirus receptor-related protein 1 [Prr1] and herpesvirus entry protein C [HveC]) was isolated. The protein encoded by this cDNA proved to be 95% identical in sequence to the human protein and to have similar herpesvirus entry activity. Upon expression of the murine cDNA in hamster cells resistant to alphaherpesvirus entry, the cells became susceptible to the entry of herpes simplex virus types 1 and 2 (HSV-1 and -2), pseudorabies virus, and bovine herpesvirus 1. HSV envelope glycoprotein D (gD), a viral ligand for human nectin-1alpha, is also a ligand for the murine homolog based on evidence that (i) a soluble hybrid protein composed in part of the murine nectin-1 ectodomain bound specifically to purified soluble forms of HSV-1 and HSV-2 gD as demonstrated by enzyme-linked immunosorbent assay, (ii) a soluble hybrid of HSV-1 gD bound to hamster cells expressing murine nectin-1alpha but not to control cells, and (iii) cells expressing both murine nectin-1alpha and one of the alphaherpesvirus gDs were resistant to entry of HSV-1, indicative of interference with entry resulting from interactions of cell-associated gD with the entry receptor. Northern blot analysis revealed that nectin-1 is expressed in most of the mouse tissues examined and at high levels in the brain, skin, and kidneys. Immunocytochemical localization demonstrated the presence of nectin-1 in epithelial cells of the mouse vagina and also in neuronal cells of the central nervous system, suggesting an expression pattern relevant to both infection at a portal of entry and spread of infection to the brain.


Subject(s)
Alphaherpesvirinae/physiology , Cell Adhesion Molecules/physiology , Receptors, Virus , Viral Envelope Proteins/physiology , Virus Replication , Amino Acid Sequence , Animals , Cattle , Cricetinae , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Nectins , Sequence Alignment , Sequence Analysis
6.
Cell Immunol ; 205(1): 24-33, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11078604

ABSTRACT

To clarify the role of tumor necrosis factor (TNF) in the inflammatory aspects of autoimmunity vs its potential role in the apoptotic elimination of autoreactive effector cells, we assessed the roles of the p55 (TNFR1/Tnfrsf1a/CD120a) and p75 (TNFR2/Tnfrsf1b/CD120b) TNF receptors in the pathogenesis of MOG(35-55)-induced experimental autoimmune encephalomyelitis (EAE). TNFR p55/p75(-/-) double knockout mice were completely resistant to clinical disease. TNFR p55(-/-) single knockout mice were also totally resistant to EAE, exhibiting reduced MOG(35-55)- specific proliferative responses and Th1 cytokine production, despite displaying equivalent DTH responses. Importantly, IL-5 was significantly increased in p55(-/-) mice. In contrast, p75(-/-) knockout mice exhibited exacerbated EAE, enhanced Th1 cytokine production, and enhanced CD4(+) and F4/80(+) CNS infiltration. Thus, p55/TNFR1 is required for the initiation of pathologic disease, whereas p75/TNFR2 may be important in regulating the immune response. These results have important implications for therapies targeting p55 and p75 receptors for treating autoimmune diseases.


Subject(s)
Antigens, CD/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Myelin-Associated Glycoprotein/immunology , Receptors, Tumor Necrosis Factor/immunology , Animals , Antigens, CD/genetics , Chemotaxis, Leukocyte , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Leukocytes, Mononuclear/immunology , Mice , Mice, Mutant Strains , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II
7.
J Virol ; 74(18): 8349-57, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954534

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.


Subject(s)
Antigens, CD/immunology , B7-1 Antigen/immunology , CD28 Antigens/immunology , Cardiovirus Infections/immunology , Demyelinating Autoimmune Diseases, CNS/immunology , Epitopes, T-Lymphocyte , Membrane Glycoproteins/immunology , Theilovirus/immunology , Animals , Antibodies/pharmacology , Antibody Formation , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen , Cardiovirus Infections/virology , Cell Division , Cytokines/metabolism , Demyelinating Autoimmune Diseases, CNS/virology , Disease Models, Animal , Female , Flow Cytometry , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred Strains , Multiple Sclerosis/immunology , Myelin Sheath/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Viral Load , Viral Plaque Assay
8.
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
9.
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
10.
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
11.
J Clin Invest ; 105(2): 223-31, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10642601

ABSTRACT

PLP139-51-induced experimental autoimmune encephalomyelitis (R-EAE) displays a relapsing-remitting paralytic course in female SJL mice. We investigated the role of apoptosis/activation-induced cell death (AICD) in the spontaneous recovery from acute disease. Clinical EAE was significantly enhanced in Fas (CD95/APO-1)-deficient SJL lpr/lpr mice, which displayed significantly increased mean peak clinical scores, reduced remission rates, and increased mortality when compared with their SJL +/lpr littermates. PLP139-151-specific proliferative responses were fairly equivalent in the 2 groups, but draining lymph node T cells from SJL lpr/lpr mice produced dramatically increased levels of IFN-gamma. Central nervous system (CNS) Fas and FasL mRNA levels in wild-type SJL (H-2(s)) mice peaked just before spontaneous disease remission and gradually declined as disease remitted. We applied the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay to detect apoptosis in situ in spinal cords of mice at various clinical stages of EAE. Most TUNEL(+) cells were found during active periods of inflammation: the acute, peak, and relapse time points. Significantly fewer apoptotic cells were observed at preclinical and remission time points. Collectively, these findings indicate that Fas-mediated apoptosis/AICD plays a major role in the spontaneous remission after the initial acute inflammatory episode and represents an important intrinsic mechanism in regulation of autoimmune responses.


Subject(s)
Apoptosis/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , fas Receptor/immunology , Animals , Chronic Disease , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Fas Ligand Protein , Female , In Situ Nick-End Labeling , Membrane Glycoproteins/genetics , Mice , Mice, Inbred Strains , Myelin Proteolipid Protein , Peptide Fragments , Polymerase Chain Reaction , RNA, Messenger/metabolism , Remission, Spontaneous , Spinal Cord/metabolism , fas Receptor/genetics
12.
J Immunol ; 164(1): 136-43, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10605004

ABSTRACT

The B7/CD28 pathway provides critical costimulatory signals required for complete T cell activation and has served as a potential target for immunotherapeutic strategies designed to regulate autoimmune diseases. This study was designed to examine the roles of CD28 and its individual ligands, B7-1 and B7-2, in experimental autoimmune encephalomyelitis (EAE), a Th1-mediated inflammatory disease of the CNS. EAE induction in CD28- or B7-deficient nonobese diabetic (NOD) mice was compared with the effects of B7/CD28 blockade using Abs in wild-type NOD mice. Disease severity was significantly reduced in CD28-deficient as well as anti-B7-1/B7-2-treated NOD mice. B7-2 appeared to play the more dominant role as there was a moderate decrease in disease incidence and severity in B7-2-deficient animals. EAE resistance was not due to the lack of effective priming of the myelin peptide-specific T cells in vivo. T cells isolated from CD28-deficient animals produced equivalent amounts of IFN-gamma and TNF-alpha in response to the immunogen, proteolipid protein 56-70. In fact, IFN-gamma and TNF-alpha production by Ag-specific T cells was enhanced in both the B7-1 and B7-2-deficient NOD mice. In contrast, peptide-specific delayed-type hypersensitivity responses in these animals were significantly decreased, suggesting a critical role for CD28 costimulation in in vivo trafficking and systemic immunity. Collectively, these results support a critical role for CD28 costimulation in EAE induction.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , B7-1 Antigen/physiology , CD28 Antigens/physiology , Encephalomyelitis, Autoimmune, Experimental/genetics , Lymphocyte Activation , Amino Acid Sequence , Animals , Antigens, CD/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-2 Antigen , CD28 Antigens/genetics , CD28 Antigens/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , Immunity, Innate , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Molecular Sequence Data , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology
13.
Blood ; 94(8): 2915-22, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10515897

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) establishes a persistent infection in the central nervous system (CNS) leading to an inflammatory demyelinating disease of the CNS in which the histology and clinical course is similar to multiple sclerosis (MS). Disease pathogenesis is primarily due to T-cell-mediated destruction of myelin, which has been attributed to cytopathic effects of the virus, but immune-mediated destruction of myelin mediated via both virus-specific and myelin-specific T cells appear to play the major role. To determine if bone marrow transplantation would be an effective therapy for a virus-initiated autoimmune disease and to better separate viral cytopathic effects from immune-mediated demyelination, we ablated the immune system of TMEV-infected animals with 1,100 cGy total body irradiation, and then the animal's immunity was reconstituted by transplantation of disease-susceptible SJL/J mice with syngeneic marrow or disease-susceptible DBA/2J with marrow from disease-resistant (C57Bl/6 x DBA/2)F1 (B6D2) donors. Hematopoietic transplant performed after onset of disease resulted in 42% mortality in SJL/J syngeneic transplants, 47% mortality in diseased DBA2 recipients restored with marrow from naive B6D2 donors, and 12% in diseased DBA2 recipients receiving marrow from B6D2 donors previously infected with TMEV. Delayed type hypersensitivity (DTH) to both virion and myelin proteins was decreased in surviving mice that underwent transplantation; however, CNS viral titers were significantly elevated compared with nontransplanted controls. We conclude that a functional immune system with appropriate T-cell responses are important in prevention of lethal cytopathic CNS effects from TMEV. Relevant to the clinical use of bone marrow transplantation, attempts to ablate the immune system in viral-mediated immune diseases or virus-initiated autoimmune disease may have acute and lethal consequences. Our results raise concern about the attempted use of autologous hematopoietic transplantation in patients with MS, an autoimmune disease with a suspected virus etiology, particularly if the graft is aggressively depleted of lymphocytes.


Subject(s)
Autoimmune Diseases/therapy , Cardiovirus Infections/therapy , Central Nervous System/virology , Demyelinating Diseases/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Lymphocyte Depletion/adverse effects , Multiple Sclerosis/therapy , Theilovirus/physiology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Contraindications , Cytokines/biosynthesis , Cytopathogenic Effect, Viral , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Disease Models, Animal , Disease Progression , Female , Genetic Predisposition to Disease , Humans , Immunocompromised Host , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred Strains , Myelin Sheath/immunology , Spinal Cord/metabolism , Spinal Cord/virology , T-Lymphocytes, Cytotoxic/immunology , Theilovirus/immunology , Theilovirus/isolation & purification , Virus Latency , Virus Replication , Whole-Body Irradiation
14.
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
15.
Acta Neuropathol ; 95(2): 136-42, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9498047

ABSTRACT

This investigation deals with the immunocytochemical localization of Cu/Zn superoxide dismutase (SOD) in the spinal cord neurons of transgenic mice that overexpress Gly93Ala mutant human Cu/Zn SOD and demonstrate clinicopathological features similar to human amyotrophic lateral sclerosis (ALS) with Cu/Zn SOD mutation. At low magnification of light microscopy, the gray and white matter of the spinal cord of Gly93Ala mice showed more intense Cu/Zn SOD immunoreactivity than that of control mice. At higher magnification, the cytoplasm of control mice neurons displayed a distinct staining for Cu/Zn SOD, whereas the surrounding neuropil was only weakly stained. In contrast, the intensity of Cu/Zn SOD immunoreactivity in the cytoplasm of the majority of Gly93Ala mice neurons was similar to that in the neuropil. Almost all neuronal hyaline inclusions (NHIs) of Gly93Ala mice were positively immunostained by antibodies to Cu/Zn SOD, ubiquitin and phosphorylated neurofilament protein (NFP), the intensities of which were much higher in the NHIs than in the surrounding cytoplasm. In control mice, significant Cu/Zn SOD precipitation was not observed to be limited to any particular region of the neuronal cytoplasm. Intracytoplasmic vacuoles in the neuronal soma and processes of Gly93Ala mice were not stained by any of these antibodies. These results indicate that Cu/Zn SOD colocalizes with ubiquitin and phosphorylated NFP in NHIs of mice expressing mutant Cu/Zn SOD; similar findings have been shown for Lewy body-like inclusions of familial ALS patients with Cu/Zn SOD mutation. Moreover, our results point to the possibility that Cu/Zn SOD mutation may have a role in the abnormal Cu/Zn SOD accumulation in the NHIs, in association with motor neuron degeneration.


Subject(s)
Hyalin/enzymology , Inclusion Bodies/enzymology , Neurons/enzymology , Spinal Cord/enzymology , Superoxide Dismutase/biosynthesis , Animals , Humans , Hyalin/ultrastructure , Inclusion Bodies/ultrastructure , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurons/cytology , Point Mutation , Spinal Cord/cytology , Superoxide Dismutase/analysis , Superoxide Dismutase/genetics
16.
J Virol ; 72(2): 1020-7, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9444995

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.


Subject(s)
Antigens, Viral/genetics , T-Lymphocytes/immunology , Theilovirus/genetics , Amino Acid Substitution , Animals , Antigen Presentation , Mice , Molecular Sequence Data , T-Lymphocytes/virology , Theilovirus/immunology , Theilovirus/pathogenicity , Virulence/genetics
17.
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
18.
Acta Neuropathol ; 93(6): 537-50, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9194892

ABSTRACT

About 15-20% of patients with familial amyotrophic lateral sclerosis (ALS) carry one of several missense mutations in the gene for Cu,Zn superoxide dismutase (SOD1). We have previously reported on an animal model of this disease produced by the transgenic expression of a mutant form of human SOD1 containing a Gly93-->Ala amino acid substitution. Several lines of transgenic mice were produced, characterized by a differing tempo and severity of disease that generally correlated with the number of mutant gene copies that these lines expressed. We reported that mice expressing high copy numbers (18-25) developed a disease with a relatively short course and with a pathology mainly characterized by severe vacuolar degeneration of motor neurons and their process. Lewy-like bodies and swollen axons were also present. The exquisite localization to motor neurons was the feature that made the pathology in these overexpressors germane to the human disease. Severe vacuolar degeneration, however, was considered to be at variance with human ALS, in which similar changes have not been described. In the present study, we have made a temporal characterization of microscopic and immunohistochemical changes in a line of transgenic mice expressing lower copy numbers of the mutant gene. These mice, designated G5/G5, survive more than 400 days and present pathological changes which are virtually identical to those in the human disease. In fact, in these animals, anterior horn cell depletion, atrophy, astrocytosis, and the presence of numerous ubiquitinated Lewy-like bodies and axonal swellings are the main pathological features, while vacuolar pathology is minimal. This study underscores the importance of the level of expression of the mutant enzyme in the resulting clinical and pathological disease, and supports the value of this transgenic model as an excellent tool for investigating both pathogenesis of human ALS and possible therapeutic avenues.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Mutation , Superoxide Dismutase/genetics , Amyotrophic Lateral Sclerosis/enzymology , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic
19.
Neuroimaging Clin N Am ; 7(2): 231-41, 1997 May.
Article in English | MEDLINE | ID: mdl-9113688

ABSTRACT

In many patients with AIDS, severe neurologic deficits develop that have been designated the cf2HIV-associated cognitive-motor complex. cf1 Pathologically, these symptoms correlate with a low-grade inflammatory condition, referred to as cf2HIV encephalitis,cf1 in which the most characteristic change is the presence of multinucleated giant cells. Cortical changes include neuronal loss and alterations of dendrites and synapses. There is pallor of white matter generally associated with the mononuclear inflammatory infiltrates. The only cells that seem to be directly infected by HIV are the microglia/monocyte and the giant cells derived from fusion of monocytes. It is hypothesized, therefore, that cortical and white matter alterations in patients with this syndrome depend on the production of injurious soluble factors liberated by these cells and by astrocytes under the influence of many of these same factors. This article reviews recent advances in the understanding of these secondary effects and discusses pathogenetic mechanisms of tissue injury.


Subject(s)
AIDS Dementia Complex/physiopathology , Central Nervous System/physiopathology , HIV Infections/physiopathology , AIDS Dementia Complex/virology , Central Nervous System/virology , HIV/physiology , HIV Infections/virology , Humans
20.
J Neurosci Res ; 45(4): 364-74, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8872896

ABSTRACT

The pathogenesis of Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease is still controversial. Our hypothesis is that primary infection of oligodendrocytes (OLGs) is not a crucial event in the pathogenesis of demyelination in this model. In fact, it has been proposed that myelin may be destroyed, as an innocent bystander, following an antiviral delayed-type hypersensitivity (DTH) response. This hypothesis would not need widespread oligodendroglial infection, because virus present in other cells would be sufficient to trigger a DTH response. The present study demonstrates that cultured OLGs and astrocytes from susceptible strains of mice (SJL and DBA) and immortalized OLGs can be infected with TMEV in vitro. Infection of OLGs, however, is at very low levels and does not result in overt cytolytic effect. In contrast, infection of immortalized OLGs is very efficient and results in clear cytolysis. Because an important characteristic of DTH responses is the liberation of potentially injurious cytokines into adjacent tissues, we also examined the effects of mouse recombinant tumor necrosis factor-alpha (TNF-alpha), interleukin-1 alpha (IL-1 alpha), and interferon-gamma (IFN-gamma) on cultured OLGs and immortalized OLGs. We found that TNF-alpha caused immortalized OLG cytotoxicity in a time- and dose-dependent manner. In contrast, no cytotoxicity was observed on primary OLGs with any of the above cytokines. To determine whether functional effects could be demonstrated on primary OLGs by either virus or cytokines, we measured mRNA expression of different myelin proteins in primary and immortalized OLGs exposed to virus or TNF-alpha. Neither the BeAn strain or the GDVII strain of TMEV interfered with myelin protein mRNA expression in primary OLGs, whereas GDVII virus dramatically reduced myelin OLG glycoprotein (MOG) mRNA in immortalized OLGs. Interestingly, although even high concentrations of TNF-alpha (10,000 U/ml) did not produce primary OLG cytotoxicity, they resulted in a significant reduction in mRNA for both myelin basic protein (MBP) and MOG in these cells. TNF-alpha (at 500 U/ml) also specifically reduced MOG mRNA in immortalized OLGs. Because immortalized OLGs are considered to be arrested at an early stage of maturation, our results suggest that immature OLGs are susceptible to both virus- and cytokine-dependent cytotoxicity, whereas mature OLGs are resistant to cytolysis by either TMEV or cytokines. TNF-alpha, however, is capable of reducing mRNA expression of myelin proteins in primary OLGs; therefore, it may participate in the induction of demyelination, as suggested by the DTH-mediated hypothesis.


Subject(s)
Astrocytes/immunology , Cytokines/pharmacology , Oligodendroglia/immunology , Theilovirus/immunology , Animals , Astrocytes/cytology , Astrocytes/virology , Autoradiography , Cell Line, Transformed/cytology , Cell Line, Transformed/immunology , Cell Line, Transformed/virology , Cells, Cultured/immunology , Cells, Cultured/virology , Cytotoxicity Tests, Immunologic , Gene Expression/physiology , Mice , Mice, Inbred DBA , Myelin Proteins/genetics , Myelin Proteins/immunology , Oligodendroglia/cytology , Oligodendroglia/virology , Polymerase Chain Reaction , Precipitin Tests , RNA, Messenger/analysis , RNA, Messenger/metabolism , RNA, Viral/analysis , Theilovirus/drug effects , Theilovirus/growth & development , Tumor Necrosis Factor-alpha/pharmacology , Viral Plaque Assay , Viral Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...