Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Transl Med ; 6(219): 219ra7, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24431111

ABSTRACT

Inflammatory monocyte-derived effector cells play an important role in the pathogenesis of numerous inflammatory diseases. However, no treatment option exists that is capable of modulating these cells specifically. We show that infused negatively charged, immune-modifying microparticles (IMPs), derived from polystyrene, microdiamonds, or biodegradable poly(lactic-co-glycolic) acid, were taken up by inflammatory monocytes, in an opsonin-independent fashion, via the macrophage receptor with collagenous structure (MARCO). Subsequently, these monocytes no longer trafficked to sites of inflammation; rather, IMP infusion caused their sequestration in the spleen through apoptotic cell clearance mechanisms and, ultimately, caspase-3-mediated apoptosis. Administration of IMPs in mouse models of myocardial infarction, experimental autoimmune encephalomyelitis, dextran sodium sulfate-induced colitis, thioglycollate-induced peritonitis, and lethal flavivirus encephalitis markedly reduced monocyte accumulation at inflammatory foci, reduced disease symptoms, and promoted tissue repair. Together, these data highlight the intricate interplay between scavenger receptors, the spleen, and inflammatory monocyte function and support the translation of IMPs for therapeutic use in diseases caused or potentiated by inflammatory monocytes.


Subject(s)
Inflammation/immunology , Inflammation/pathology , Microspheres , Monocytes/immunology , Animals , Apoptosis , Brain/pathology , Cell Movement , Cell Survival , Colitis/pathology , Colitis/prevention & control , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Kidney/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Myocardium/pathology , Particle Size , Peritoneum/pathology , Polystyrenes/chemistry , Receptors, Immunologic/metabolism , Reperfusion Injury/prevention & control , Spleen/pathology , West Nile Fever
2.
J Neuroinflammation ; 9: 246, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23111065

ABSTRACT

Infiltration of Ly6C(hi) monocytes from the blood is a hallmark of viral encephalitis. In mice with lethal encephalitis caused by West Nile virus (WNV), an emerging neurotropic flavivirus, inhibition of Ly6C(hi) monocyte trafficking into the brain by anti-very late antigen (VLA)-4 integrin antibody blockade at the time of first weight loss and leukocyte influx resulted in long-term survival of up to 60% of infected mice, with subsequent sterilizing immunity. This treatment had no effect on viral titers but appeared to be due to inhibition of Ly6C(hi) macrophage immigration. Although macrophages isolated from the infected brain induced WNV-specific CD4(+) T-cell proliferation, T cells did not directly contribute to pathology, but are likely to be important in viral control, as antibody-mediated T-cell depletion could not reproduce the therapeutic benefit of anti-VLA-4. Instead, 70% of infiltrating inflammatory monocyte-derived macrophages were found to be making nitric oxide (NO). Furthermore, aminoguanidine-mediated inhibition of induced NO synthase activity in infiltrating macrophages significantly prolonged survival, indicating involvement of NO in the immunopathology. These data show for the first time the therapeutic effects of temporally targeting pathogenic NO-producing macrophages during neurotropic viral encephalitis.


Subject(s)
Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Macrophages/immunology , Macrophages/metabolism , West Nile Fever , Animals , Antigens, CD/metabolism , Brain/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Viral/physiology , Glial Fibrillary Acidic Protein/metabolism , Integrins/genetics , Integrins/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/immunology , Nitric Oxide Synthase Type II , West Nile Fever/immunology , West Nile Fever/metabolism , West Nile Fever/pathology
3.
Nat Biotechnol ; 30(12): 1217-24, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23159881

ABSTRACT

Aberrant T-cell activation underlies many autoimmune disorders, yet most attempts to induce T-cell tolerance have failed. Building on previous strategies for tolerance induction that exploited natural mechanisms for clearing apoptotic debris, we show that antigen-decorated microparticles (500-nm diameter) induce long-term T-cell tolerance in mice with relapsing experimental autoimmune encephalomyelitis. Specifically, intravenous infusion of either polystyrene or biodegradable poly(lactide-co-glycolide) microparticles bearing encephalitogenic peptides prevents the onset and modifies the course of the disease. These beneficial effects require microparticle uptake by marginal zone macrophages expressing the scavenger receptor MARCO and are mediated in part by the activity of regulatory T cells, abortive T-cell activation and T-cell anergy. Together these data highlight the potential for using microparticles to target natural apoptotic clearance pathways to inactivate pathogenic T cells and halt the disease process in autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immune Tolerance , Myelin Proteolipid Protein/administration & dosage , Myelin Proteolipid Protein/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/administration & dosage , Biotechnology , Clonal Anergy , Female , Infusions, Intravenous , Interleukin-10/immunology , Lymphocyte Activation , Mice , Microspheres , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Polyglactin 910 , Polystyrenes , T-Lymphocytes, Regulatory/immunology
4.
J Immunol ; 187(5): 2405-17, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21821796

ABSTRACT

Ag-specific tolerance is a highly desired therapy for immune-mediated diseases. Intravenous infusion of protein/peptide Ags linked to syngeneic splenic leukocytes with ethylene carbodiimide (Ag-coupled splenocytes [Ag-SP]) has been demonstrated to be a highly efficient method for inducing peripheral, Ag-specific T cell tolerance for treatment of autoimmune disease. However, little is understood about the mechanisms underlying this therapy. In this study, we show that apoptotic Ag-SP accumulate in the splenic marginal zone, where their uptake by F4/80(+) macrophages induces production of IL-10, which upregulates the expression of the immunomodulatory costimulatory molecule PD-L1 that is essential for Ag-SP tolerance induction. Ag-SP infusion also induces T regulatory cells that are dispensable for tolerance induction but required for long-term tolerance maintenance. Collectively, these results indicate that Ag-SP tolerance recapitulates how tolerance is normally maintained in the hematopoietic compartment and highlight the interplay between the innate and adaptive immune systems in the induction of Ag-SP tolerance. To our knowledge, we show for the first time that tolerance results from the synergistic effects of two distinct mechanisms, PD-L1-dependent T cell-intrinsic unresponsiveness and the activation of T regulatory cells. These findings are particularly relevant as this tolerance protocol is currently being tested in a Phase I/IIa clinical trial in new-onset relapsing-remitting multiple sclerosis.


Subject(s)
Immune Tolerance/immunology , Macrophages/immunology , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Apoptosis/immunology , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , B7-H1 Antigen , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunohistochemistry , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Macrophage Activation/immunology , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Peptides/immunology , Spleen/cytology
5.
Immunotherapy ; 3(7): 853-70, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21751954

ABSTRACT

In recent years, substantial advances in T-cell immunosuppressive strategies and their translation to routine clinical practice have revolutionized management and outcomes in autoimmune disease and solid organ transplantation. More than 80 diseases have been considered to have an autoimmune etiology, such that autoimmune-associated morbidity and mortality rank as third highest in developed countries, after cardiovascular diseases and cancer. Solid organ transplantation has become the therapy of choice for many end-stage organ diseases. Short-term outcomes such as patient and allograft survival at 1 year, acute rejection rates, as well as time course of disease progression and symptom control have steadily improved. However, despite the use of newer immunosuppressive drug combinations, improvements in long-term allograft survival and complete resolution of autoimmunity remain elusive. In addition, the chronic use of nonspecifically targeted immunosuppressive drugs is associated with significant adverse effects and increased morbidity and mortality. In this article, we discuss the current clinical tools for immune suppression and attempts to induce long-term T-cell tolerance induction as well as much-needed future approaches to produce more short-acting, antigen-specific agents, which may optimize outcomes in the clinic.


Subject(s)
Autoimmune Diseases , Hematopoietic Stem Cell Transplantation/methods , Organ Transplantation/methods , T-Lymphocytes , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Calcineurin Inhibitors , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Humans , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Janus Kinase 3/antagonists & inhibitors , Piperidines , Protein Kinase C/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation Immunology/immunology , Transplantation Tolerance/immunology
6.
J Autoimmun ; 36(2): 142-54, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21273044

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) serves as virus-induced model of chronic progressive multiple sclerosis. Infection of susceptible SJL/J mice leads to life-long CNS virus persistence and a progressive autoimmune demyelinating disease mediated by myelin-specific T cells activated via epitope spreading. In contrast, virus is rapidly cleared by a robust CTL response in TMEV-IDD-resistant C57BL/6 mice. We investigated whether differential induction of regulatory T cells (Tregs) controls susceptibility to TMEV-IDD. Infection of disease-susceptible SJL/J, but not B6 mice, leads to rapid activation and expansion of Tregs resulting in an unfavorable CNS ratio of Treg:Teffector cells. In addition, anti-CD25-induced inactivation of Tregs in susceptible SJL/J, but not resistant B6, mice results in significantly decreased clinical disease concomitant with enhanced anti-viral CD4(+), CD8(+) and antibody responses resulting in decreased CNS viral titers. This is the first demonstration that virus-induced Treg activation regulates susceptibility to autoimmune disease differentially in susceptible and resistant strains of mice and provides a new mechanistic explanation for the etiology of infection-induced autoimmunity.


Subject(s)
Demyelinating Diseases/immunology , Disease Models, Animal , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/immunology , Theilovirus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cardiovirus Infections/immunology , Cardiovirus Infections/metabolism , Cardiovirus Infections/pathology , Cardiovirus Infections/prevention & control , Cell Proliferation , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Demyelinating Diseases/prevention & control , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Glucocorticoid-Induced TNFR-Related Protein , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/immunology , Severity of Illness Index , T-Lymphocytes, Regulatory/metabolism , Theilovirus/drug effects
7.
Virology ; 402(1): 102-11, 2010 Jun 20.
Article in English | MEDLINE | ID: mdl-20381109

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) is a relevant mouse model of multiple sclerosis. Infection of susceptible SJL/J mice leads to life-long CNS virus persistence and development of a chronic T cell-mediated autoimmune demyelinating disease triggered via epitope spreading to endogenous myelin epitopes. Potent CNS-infiltrating CD8(+) T cell responses to TMEV epitopes have previously been shown to be induced in both disease-susceptible SJL/J and resistant C57BL/6 mice, in which the virus is rapidly cleared. Specific tolerization of SJL CD8(+) T cells specific for the immunodominant TMEV VP3(159)(-)(166) epitope has no effect on viral load or development of clinical TMEV-IDD, but adoptive transfer of activated CD8(+) VP3(159)(-)(166)-specific T cell blasts shortly after TMEV infection to boost the early anti-viral response leads to clearance of CNS virus and protection from subsequent TMEV-IDD. These studies have important implications for vaccine strategies and treatment of chronic infections in humans.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cardiovirus Infections/veterinary , Demyelinating Diseases/prevention & control , T-Lymphocytes, Cytotoxic/immunology , Theilovirus/immunology , Adoptive Transfer , Animals , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Epitopes, T-Lymphocyte/immunology , Mice , Viral Load
8.
Nat Rev Immunol ; 9(4): 246-58, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19319143

ABSTRACT

The predisposition of individuals to several common autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis, is genetically linked to certain human MHC class II molecules and other immune modulators. However, genetic predisposition is only one risk factor for the development of these diseases, and low concordance rates in monozygotic twins, as well as the geographical distribution of disease risk, suggest the involvement of environmental factors in the development of these diseases. Among these environmental factors, infections have been implicated in the onset and/or promotion of autoimmunity. In this Review, we outline the mechanisms by which viral infection can trigger autoimmune disease and describe the pathways by which infection and immune control of infectious disease might be dysregulated during autoimmunity.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmunity/immunology , Virus Diseases/complications , Virus Diseases/immunology , Adjuvants, Immunologic/physiology , Animals , Disease Models, Animal , Humans , Models, Immunological , Molecular Mimicry/immunology
9.
J Exp Med ; 205(10): 2319-37, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18779347

ABSTRACT

In a lethal West Nile virus (WNV) model, central nervous system infection triggered a threefold increase in CD45(int)/CD11b(+)/CD11c(-) microglia at days 6-7 postinfection (p.i.). Few microglia were proliferating, suggesting that the increased numbers were derived from a migratory precursor cell. Depletion of "circulating" (Gr1(-)(Ly6C(lo))CX3CR1(+)) and "inflammatory" (Gr1(hi)/Ly6C(hi)/CCR2(+)) classical monocytes during infection abrogated the increase in microglia. C57BL/6 chimeras reconstituted with cFMS-enhanced green fluorescent protein (EGFP) bone marrow (BM) showed large numbers of peripherally derived (GFP(+)) microglia expressing GR1(+)(Ly6C(+)) at day 7 p.i., suggesting that the inflammatory monocyte is a microglial precursor. This was confirmed by adoptive transfer of labeled BM (Ly6C(hi)/CD115(+)) or circulating inflammatory monocytes that trafficked to the WNV-infected brain and expressed a microglial phenotype. CCL2 is a chemokine that is highly expressed during WNV infection and important in inflammatory monocyte trafficking. Neutralization of CCL2 not only reduced the number of GFP(+) microglia in the brain during WNV infection but prolonged the life of infected animals. Therefore, CCL2-dependent inflammatory monocyte migration is critical for increases in microglia during WNV infection and may also play a pathogenic role during WNV encephalitis.


Subject(s)
Antigens, Ly/immunology , Brain/immunology , Brain/virology , Microglia/immunology , Monocytes/immunology , Stem Cells/immunology , West Nile Fever/immunology , Adoptive Transfer , Animals , Blood-Brain Barrier/physiology , Brain/cytology , Brain/pathology , Chemokine CCL2/immunology , Clodronic Acid/metabolism , Female , Leukocyte Common Antigens/immunology , Liposomes/chemistry , Liposomes/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/physiology , Monocytes/cytology , Stem Cells/cytology , Survival Rate , West Nile Fever/pathology , West Nile virus
10.
Virology ; 375(1): 24-36, 2008 May 25.
Article in English | MEDLINE | ID: mdl-18289626

ABSTRACT

Intracerebral infection of susceptible strains of mice, e.g. SJL/J, with Theiler's murine encephalomyelitis virus (TMEV) leads to a persistent CNS infection accompanied by development of a chronic-progressive inflammatory CNS autoimmune demyelinating disease which is clinically and pathologically similar to human multiple sclerosis. In contrast, resistant strains of mice, e.g. C57BL/6 (B6), effectively clear TMEV from the CNS and do not develop demyelinating disease. Although CD8(+) T cells are crucial for viral clearance in B6 mice, SJL mice also mount potent CD8(+) T cell responses against virus, thus the reason for the viral persistence in the CNS in these mice is unclear. Here, we examined innate anti-viral responses of CNS-resident astrocytes as a potential determinant of viral persistence and disease susceptibility. We demonstrate that B6 astrocytes produce significantly higher levels of cytokines, chemokines and adhesion molecules in response to TMEV infection, or stimulation with IFN-gamma and TNF-alpha or poly I:C than SJL mice. In addition, TMEV more effectively induces MHC I molecules on B6 astrocytes than SJL, corresponding with an increased ability to activate TMEV-specific CD8(+) T cells directly ex vivo. These results suggest that enhanced anti-viral responses of B6 astrocytes contribute to the ability of these mice to clear TMEV from the CNS and therefore to their resistance to the development of autoimmune demyelinating disease.


Subject(s)
Astrocytes/immunology , Demyelinating Diseases/immunology , Demyelinating Diseases/virology , Theilovirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/biosynthesis , Cells, Cultured , Cytokines/biosynthesis , Female , Flow Cytometry , Histocompatibility Antigens Class I/biosynthesis , Interferon-gamma/immunology , Mice , Mice, Inbred C57BL , Poly I-C/immunology , Pregnancy , Tumor Necrosis Factor-alpha/immunology
11.
Therapy ; 5(5): 631-640, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-20357912

ABSTRACT

Multiple Sclerosis (MS) is an inflammatory demyelinating disorder of the central nervous system (CNS). Since current treatments are aimed at nonspecifically down-regulating inflammation and natural mechanisms of repair and remyelination within the CNS are inadequate for recovery of function, MS patients presently only have available treatments that delay symptom progression. The complex nature of this disease means that only multifaceted treatments hold the promise of a cure. Recent studies indicate that the ER stress response, a cellular pathway that allows a cell to survive and recover from a stressful event, could be elicited to help the myelin-generating and myelin-support cells of the CNS survive inflammatory insult.

12.
J Neurochem ; 103(3): 1019-30, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17854352

ABSTRACT

Seizures are a major complication of viral encephalitis. However, the mechanisms of seizure-associated neuronal dysfunction remain poorly understood. We report that intranasal inoculation with West Nile virus (WNV) (Sarafend) causes limbic seizures in C57BL/6 mice, but not in interferon (IFN)-gamma-deficient (IFN-gamma-/-) mice. Both strains showed similar levels of virus in the brain, as well as similar concentrations of the cytokines, tumor necrosis factor and interleukin-6, both of which can alter neuronal excitability. Experiments in chimeric IFN-gamma-/- mice reconstituted with IFN-gamma-producing leukocytes showed that IFN-gamma is not required during central nervous system infection for limbic seizure development, suggesting a role for IFN-gamma in the developing brain. This was supported responses to pentylenetetrazole, kainic acid (KA), and N-methyl-d-aspartate (NMDA). Both strains of mice exhibited similar behavior after pentylenetetrazole challenge. However, while NMDA and KA treatment resulted in characteristic seizures in C57BL/6 mice, these responses were diminished (NMDA treatment) or absent (KA treatment) in IFN-gamma-/- mice. Furthermore, NMDA-receptor blockade with MK-801 in WNV-infected C57BL/6 mice abrogated seizures and prolonged survival. Our data show that IFN-gamma plays an important role in the development of the excitatory seizure pathways in the brain and that these cascades become pathogenic in encephalitic WNV infection.


Subject(s)
Brain/immunology , Brain/virology , Interferon-gamma/immunology , Seizures/immunology , Seizures/virology , West Nile Fever/immunology , Animals , Brain/physiopathology , Chlorocebus aethiops , Convulsants/pharmacology , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Female , Genetic Predisposition to Disease/genetics , Glutamic Acid/metabolism , Interferon-gamma/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Limbic Encephalitis/genetics , Limbic Encephalitis/immunology , Limbic Encephalitis/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/immunology , Neural Pathways/physiopathology , Neural Pathways/virology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/genetics , Synaptic Transmission/genetics , Synaptic Transmission/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vero Cells , West Nile Fever/complications , West Nile Fever/physiopathology
13.
J Virol ; 81(12): 6584-93, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17428853

ABSTRACT

Tolerance induced by the intravenous injection of peptide-pulsed, ethylene carbodiimide (ECDI)-fixed splenic antigen-presenting cells (Ag-SP) is a safe and effective method of inducing specific unresponsiveness in CD4+ T cells for the prevention and treatment of a variety of autoimmune diseases. We determined whether Ag-SP tolerance could also be used to tolerize CD8+ T cells. We show in the Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease model of multiple sclerosis that CD8+ T cells specific for both dominant and subdominant epitopes can be rendered tolerant. Interestingly, although virus clearance was delayed, lack of the virus-specific cytotoxic T-lymphocyte response did not result in the conversion of normally TMEV-resistant C57BL/6 mice to a susceptible phenotype. Importantly, we found that Ag-SP tolerance may not be a practical treatment for human diseases in which CD8+ T cells play a major role in pathogenesis, as tolerance induction in mice previously infected with TMEV led to a severe, often fatal reaction.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/chemistry , Immune Tolerance , Theilovirus/genetics , Animals , Antigen-Presenting Cells/metabolism , Autoimmune Diseases/metabolism , Carbodiimides/chemistry , Central Nervous System/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Spleen/metabolism , Theilovirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL