Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Nanomedicine ; 40: 102481, 2022 02.
Article in English | MEDLINE | ID: mdl-34748963

ABSTRACT

Tolerance induction is central to the suppression of autoimmunity. Here, we engineered the preferential uptake of nano-conjugated autoantigens by spleen-resident macrophages to re-introduce self-tolerance and suppress autoimmunity. The brain autoantigen, myelin oligodendrocyte glycoprotein (MOG), was conjugated to 200 or 500 nm silica nanoparticles (SNP) and delivered to the spleen and liver-resident macrophages of experimental autoimmune encephalomyelitis (EAE) mice, used as a model of multiple sclerosis. MOG-SNP conjugates significantly reduced signs of EAE at a very low dose (50 µg) compared to the higher dose (>800 µg) of free-MOG. This was associated with reduced proliferation of splenocytes and pro-inflammatory cytokines secretion, decreased spinal cord inflammation, demyelination and axonal damage. Notably, biodegradable porous SNP showed an enhanced disease suppression assisted by elevated levels of regulatory T cells and programmed-death ligands (PD-L1/2) in splenic and lymph node cells. Our results demonstrate that targeting nano-conjugated autoantigens to tissue-resident macrophages in lymphoid organs can effectively suppress autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Nanoparticles , Animals , Autoimmunity , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Myelin-Oligodendrocyte Glycoprotein/therapeutic use
2.
Proc Natl Acad Sci U S A ; 115(39): 9773-9778, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30194232

ABSTRACT

The anti-CD20 antibody ocrelizumab, approved for treatment of multiple sclerosis, leads to rapid elimination of B cells from the blood. The extent of B cell depletion and kinetics of their recovery in different immune compartments is largely unknown. Here, we studied how anti-CD20 treatment influences B cells in bone marrow, blood, lymph nodes, and spleen in models of experimental autoimmune encephalomyelitis (EAE). Anti-CD20 reduced mature B cells in all compartments examined, although a subpopulation of antigen-experienced B cells persisted in splenic follicles. Upon treatment cessation, CD20+ B cells simultaneously repopulated in bone marrow and spleen before their reappearance in blood. In EAE induced by native myelin oligodendrocyte glycoprotein (MOG), a model in which B cells are activated, B cell recovery was characterized by expansion of mature, differentiated cells containing a high frequency of myelin-reactive B cells with restricted B cell receptor gene diversity. Those B cells served as efficient antigen-presenting cells (APCs) for activation of myelin-specific T cells. In MOG peptide-induced EAE, a purely T cell-mediated model that does not require B cells, in contrast, reconstituting B cells exhibited a naive phenotype without efficient APC capacity. Our results demonstrate that distinct subpopulations of B cells differ in their sensitivity to anti-CD20 treatment and suggest that differentiated B cells persisting in secondary lymphoid organs contribute to the recovering B cell pool.


Subject(s)
Antigens, CD20/immunology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Bone Marrow Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Myelin Sheath/immunology , Spleen/cytology , Spleen/immunology
3.
Ann Neurol ; 77(5): 902-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25712734

ABSTRACT

Natalizumab, which binds very late antigen-4 (VLA-4), is a potent therapy for multiple sclerosis (MS). Studies have focused primarily upon its capacity to interfere with T-cell migration into the central nervous system (CNS). B cells are important in MS pathogenesis and express high levels of VLA-4. Here, we report that the selective inhibition of VLA-4 expression on B cells impedes CNS accumulation of B cells, and recruitment of Th17 cells and macrophages, and reduces susceptibility to experimental autoimmune encephalomyelitis. These results underscore the importance of B-cell VLA-4 expression in the pathogenesis of CNS autoimmunity and provide insight regarding mechanisms that may contribute to the benefit of natalizumab in MS, as well as candidate therapeutics that selectively target B cells.


Subject(s)
Autoimmunity/immunology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4beta1/deficiency , Animals , B-Lymphocytes/metabolism , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Th17 Cells/immunology , Th17 Cells/metabolism
4.
J Neuroinflammation ; 12: 112, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26036872

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system (CNS). In recent years, it has been found that cells such as human amnion epithelial cells (hAECs) have the ability to modulate immune responses in vitro and in vivo and can differentiate into multiple cell lineages. Accordingly, we investigated the immunoregulatory effects of hAECs as a potential therapy in an MS-like disease, EAE (experimental autoimmune encephalomyelitis), in mice. METHODS: Using flow cytometry, the phenotypic profile of hAECs from different donors was assessed. The immunomodulatory properties of hAECs were examined in vitro using antigen-specific and one-way mixed lymphocyte proliferation assays. The therapeutic efficacy of hAECs was examined using a relapsing-remitting model of EAE in NOD/Lt mice. T cell responsiveness, cytokine secretion, T regulatory, and T helper cell phenotype were determined in the peripheral lymphoid organs and CNS of these animals. RESULTS: In vitro, hAECs suppressed both specific and non-specific T cell proliferation, decreased pro-inflammatory cytokine production, and inhibited the activation of stimulated T cells. Furthermore, T cells retained their naïve phenotype when co-cultured with hAECs. In vivo studies revealed that hAECs not only suppressed the development of EAE but also prevented disease relapse in these mice. T cell responses and production of the pro-inflammatory cytokine interleukin (IL)-17A were reduced in hAEC-treated mice, and this was coupled with a significant increase in the number of peripheral T regulatory cells and naïve CD4+ T cells. Furthermore, increased proportions of Th2 cells in the peripheral lymphoid organs and within the CNS were observed. CONCLUSION: The therapeutic effect of hAECs is in part mediated by inducing an anti-inflammatory response within the CNS, demonstrating that hAECs hold promise for the treatment of autoimmune diseases like MS.


Subject(s)
Amnion/cytology , Amnion/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Epithelial Cells/cytology , Epithelial Cells/immunology , Immunosuppression Therapy/methods , Amnion/transplantation , Animals , Cell Proliferation/physiology , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Epithelial Cells/transplantation , Female , Humans , In Vitro Techniques , Lymphoid Tissue/pathology , Mice , Mice, Inbred NOD , Phenotype , T-Lymphocytes/pathology , T-Lymphocytes, Regulatory/pathology
5.
Biol Chem ; 396(8): 923-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25719317

ABSTRACT

Induced pluripotent stem cell (iPSC)-derived neurospheres, which consist mainly of neural progenitors, are considered to be a good source of neural cells for transplantation in regenerative medicine. In this study, we have used lithium chloride, which is known to be a neuroprotective agent, in an iPSC-derived neurosphere model, and examined both the formation rate and size of the neurospheres as well as the proliferative and apoptotic status of their contents. Our results showed that lithium enhanced the formation and the sizes of the iPSC-derived neurospheres, increased the number of Ki67-positive proliferating cells, but reduced the number of the TUNEL-positive apoptotic cells. This increased number of Ki67 proliferating cells was secondary to the decreased apoptosis and not to the stimulation of cell cycle entry, as the expression of the proliferation marker cyclin D1 mRNA did not change after lithium treatment. Altogether, we suggest that lithium enhances the survival of neural progenitors and thus the quality of the iPSC-derived neurospheres, which may strengthen the prospect of using lithium-treated pluripotent cells and their derivatives in a clinical setting.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Lithium Chloride/pharmacology , Neurons/drug effects , Apoptosis/drug effects , Cells, Cultured , Cyclin D1/genetics , Humans , In Situ Nick-End Labeling , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics
6.
J Proteome Res ; 13(8): 3655-70, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24933266

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a murine model of multiple sclerosis, a chronic neurodegenerative and inflammatory autoimmune condition of the central nervous system (CNS). Pathology is driven by the infiltration of autoreactive CD4(+) lymphocytes into the CNS, where they attack neuronal sheaths causing ascending paralysis. We used an isotope-coded protein labeling approach to investigate the proteome of CD4(+) cells isolated from the spinal cord and brain of mice at various stages of EAE progression in two EAE disease models: PLP139-151-induced relapsing-remitting EAE and MOG35-55-induced chronic EAE, which emulate the two forms of human multiple sclerosis. A total of 1120 proteins were quantified across disease onset, peak-disease, and remission phases of disease, and of these 13 up-regulated proteins of interest were identified with functions relating to the regulation of inflammation, leukocyte adhesion and migration, tissue repair, and the regulation of transcription/translation. Proteins implicated in processes such as inflammation (S100A4 and S100A9) and tissue repair (annexin A1), which represent key events during EAE progression, were validated by quantitative PCR. This is the first targeted analysis of autoreactive cells purified from the CNS during EAE, highlighting fundamental CD4(+) cell-driven processes that occur during the initiation of relapse and remission stages of disease.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Central Nervous System/cytology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation, Neoplastic/genetics , Proteome/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion/genetics , Cell Movement/genetics , Central Nervous System/metabolism , Chromatography, High Pressure Liquid , Female , Flow Cytometry , Mass Spectrometry , Mice , Molecular Sequence Data , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/immunology , Myelin-Oligodendrocyte Glycoprotein/genetics , Peptide Fragments/genetics , Peptide Fragments/immunology , Pertussis Toxin , Proteome/genetics
7.
Brain Behav Immun ; 30: 103-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23369732

ABSTRACT

Interleukin (IL)-10 is an important immunoregulatory cytokine shown to impact inflammatory processes as manifested in patients with multiple sclerosis (MS) and in its animal model, experimental autoimmune encephalomyelitis (EAE). Several lines of evidence indicate that the effectiveness of IL-10-based therapies may be dependent on the timing and mode of delivery. In the present study we engineered the expression of IL-10 in human adipose-derived mesenchymal stem cells (Adi-IL-10-MSCs) and transplanted these cells early in the disease course to mice with EAE. Adi-IL-10-MSCs transplanted via the intraperitoneal route prevented or delayed the development of EAE. This protective effect was associated with several anti-inflammatory response mechanisms, including a reduction in peripheral T-cell proliferative responses, a decrease in pro-inflammatory cytokine secretion as well as a preferential inhibition of Th17-mediated neuroinflammation. In vitro analyses revealed that Adi-IL-10-MSCs inhibited the phenotypic maturation, cytokine production and antigen presenting capacity of bone marrow-derived myeloid dendritic cells, suggesting that the mechanism of action may involve an indirect effect on pathogenic T-cells via the modulation of antigen presenting cell function. Collectively, these results suggest that early intervention with gene modified Adi-MSCs may be beneficial for the treatment of autoimmune diseases such as MS.


Subject(s)
Adipocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Interleukin-10/metabolism , Mesenchymal Stem Cells/metabolism , Adipocytes/transplantation , Animals , Autoimmunity/immunology , Cell Differentiation/immunology , Cell Proliferation , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Mesenchymal Stem Cell Transplantation/methods , Mice , T-Lymphocytes/immunology
8.
Brain ; 135(Pt 6): 1794-818, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22544872

ABSTRACT

Multiple sclerosis involves demyelination and axonal degeneration of the central nervous system. The molecular mechanisms of axonal degeneration are relatively unexplored in both multiple sclerosis and its mouse model, experimental autoimmune encephalomyelitis. We previously reported that targeting the axonal growth inhibitor, Nogo-A, may protect against neurodegeneration in experimental autoimmune encephalomyelitis; however, the mechanism by which this occurs is unclear. We now show that the collapsin response mediator protein 2 (CRMP-2), an important tubulin-associated protein that regulates axonal growth, is phosphorylated and hence inhibited during the progression of experimental autoimmune encephalomyelitis in degenerating axons. The phosphorylated form of CRMP-2 (pThr555CRMP-2) is localized to spinal cord neurons and axons in chronic-active multiple sclerosis lesions. Specifically, pThr555CRMP-2 is implicated to be Nogo-66 receptor 1 (NgR1)-dependent, since myelin oligodendrocyte glycoprotein (MOG)(35-55)-induced NgR1 knock-out (ngr1(-)(/)(-)) mice display a reduced experimental autoimmune encephalomyelitis disease progression, without a deregulation of ngr1(-)(/)(-) MOG(35-55)-reactive lymphocytes and monocytes. The limitation of axonal degeneration/loss in experimental autoimmune encephalomyelitis-induced ngr1(-)(/)(-) mice is associated with lower levels of pThr555CRMP-2 in the spinal cord and optic nerve during experimental autoimmune encephalomyelitis. Furthermore, transduction of retinal ganglion cells with an adeno-associated viral vector encoding a site-specific mutant T555ACRMP-2 construct, limits optic nerve axonal degeneration occurring at peak stage of experimental autoimmune encephalomyelitis. Therapeutic administration of the anti-Nogo(623-640) antibody during the course of experimental autoimmune encephalomyelitis, associated with an improved clinical outcome, is demonstrated to abrogate the protein levels of pThr555CRMP-2 in the spinal cord and improve pathological outcome. We conclude that phosphorylation of CRMP-2 may be downstream of NgR1 activation and play a role in axonal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis. Blockade of Nogo-A/NgR1 interaction may serve as a viable therapeutic target in multiple sclerosis.


Subject(s)
Axons/metabolism , Encephalomyelitis, Autoimmune, Experimental/complications , Intercellular Signaling Peptides and Proteins/metabolism , Multiple Sclerosis/pathology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Adult , Analysis of Variance , Animals , Antibodies/therapeutic use , Axons/pathology , Axons/ultrastructure , CD3 Complex/metabolism , Cell Line, Tumor , Demyelinating Diseases/etiology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/immunology , Gene Expression Regulation/genetics , Glycoproteins/adverse effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Multiple Sclerosis/complications , Mutation/genetics , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/deficiency , Myelin Proteins/immunology , Myelin-Oligodendrocyte Glycoprotein , Nerve Degeneration/etiology , Nerve Tissue Proteins/genetics , Neuroblastoma/pathology , Neurofilament Proteins/metabolism , Nogo Receptor 1 , Optic Nerve/metabolism , Optic Nerve/pathology , Peptide Fragments/adverse effects , Phosphorylation , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/immunology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
9.
Mol Cell Proteomics ; 10(6): M110.000042, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21421798

ABSTRACT

Neurodegenerative diseases, such as multiple sclerosis represent global health issues. Accordingly, there is an urgent need to understand the pathogenesis of this and other central nervous system disorders, so that more effective therapeutics can be developed. Cerebrospinal fluid is a potential source of important reporter molecules released from various cell types as a result of central nervous system pathology. Here, we report the development of an unbiased approach for the detection of reactive cerebrospinal fluid molecules and target brain proteins from patients with multiple sclerosis. To help identify molecules that may serve as clinical biomarkers for multiple sclerosis, we have biotinylated proteins present in the cerebrospinal fluid and tested their reactivity against brain homogenate as well as myelin and myelin-axolemmal complexes. Proteins were separated by two-dimensional gel electrophoresis, blotted onto membranes and probed separately with biotinylated unprocessed cerebrospinal fluid samples. Protein spots that reacted to two or more multiple sclerosis-cerebrospinal fluids were further analyzed by matrix assisted laser desorption ionization-time-of-flight time-of-flight mass spectrometry. In addition to previously reported proteins found in multiple sclerosis cerebrospinal fluid, such as αß crystallin, enolase, and 14-3-3-protein, we have identified several additional molecules involved in mitochondrial and energy metabolism, myelin gene expression and/or cytoskeletal organization. These include aspartate aminotransferase, cyclophilin-A, quaking protein, collapsin response mediator protein-2, ubiquitin carboxy-terminal hydrolase L1, and cofilin. To further validate these findings, the cellular expression pattern of collapsin response mediator protein-2 and ubiquitin carboxy-terminal hydrolase L1 were investigated in human chronic-active MS lesions by immunohistochemistry. The observation that in multiple sclerosis lesions phosphorylated collapsin response mediator protein-2 was increased, whereas Ubiquitin carboxy-terminal hydrolase L1 was down-regulated, not only highlights the importance of these molecules in the pathology of this disease, but also illustrates the use of our approach in attempting to decipher the complex pathological processes leading to multiple sclerosis and other neurodegenerative diseases.


Subject(s)
Cerebrospinal Fluid Proteins/chemistry , Multiple Sclerosis/cerebrospinal fluid , Adult , Aged , Axons/metabolism , Biotinylation , Blotting, Western , Brain/immunology , Brain/metabolism , Brain/pathology , Cerebrospinal Fluid Proteins/immunology , Cerebrospinal Fluid Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Energy Metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Myelin Sheath/enzymology , Myelin Sheath/immunology , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Proteomics , Ubiquitin Thiolesterase/metabolism
10.
Neuroimage ; 59(4): 3624-40, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22119649

ABSTRACT

In the last two decades the field of infrared spectroscopy has seen enormous advances in both instrumentation and the development of bioinformatic methods for spectral analysis, allowing the examination of a large variety of healthy and diseased samples, including biological fluids, isolated cells, whole tissues, and tissue sections. The non-destructive nature of the technique, together with the ability to directly probe biochemical changes without the addition of stains or contrast agents, enables a range of complementary analyses. This review focuses on the application of Fourier transform infrared (FTIR) microspectroscopy to analyse central nervous system tissues, with the aim of understanding the biochemical and structural changes associated with neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, transmissible spongiform encephalopathies, multiple sclerosis, as well as brain tumours. Modern biospectroscopic methods that combine FTIR microspectroscopy with bioinformatic analysis constitute a powerful new methodology that can discriminate pathology from normal healthy tissue in a rapid, unbiased fashion, with high sensitivity and specificity. Notably, the ability to detect protein secondary structural changes associated with Alzheimer's plaques, neurons in Parkinson's disease, and in some spectra from meningioma, as well as in the animal models of Alzheimer's disease, transmissible spongiform encephalopathies, and multiple sclerosis, illustrates the power of this technology. The capacity to offer insight into the biochemical and structural changes underpinning aetio-pathogenesis of diseases in tissues provides both a platform to investigate early pathologies occurring in a variety of experimentally induced and naturally occurring central nervous system diseases, and the potential to evaluate new therapeutic approaches.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Spectroscopy, Fourier Transform Infrared/methods , Humans
11.
J Neurosci Res ; 89(5): 639-49, 2011 May.
Article in English | MEDLINE | ID: mdl-21344476

ABSTRACT

Demyelination coincides with numerous changes of gene expression in the central nervous system (CNS). Cystatin F, which is a papain-like lysosomal cysteine proteinase inhibitor that is normally expressed by immune cells and not in the brain, is massively induced in the CNS during acute demyelination. We found that microglia, which are monocyte/macrophage-lineage cells in the CNS, express cystatin F only during demyelination. By using several demyelinating animal models and the spinal cord tissues from multiple sclerosis (MS) patients, we examined spatiotemporal expression pattern of cystatin F by in situ hybridization and immunohistochemistry. We found that the timing of cystatin F induction matches with ongoing demyelination, and the places with cystatin F expression overlapped with the remyelinating area. Most interestingly, cystatin F induction ceased in chronic demyelination, in which remyelinating ability was lost. These findings demonstrate that the expression of cystatin F indicates the occurrence of ongoing demyelination/remyelination and the absence of cystatin F expression indicates the cessation of remyelination in the demyelinating area.


Subject(s)
Cystatins/biosynthesis , Demyelinating Autoimmune Diseases, CNS/metabolism , Microglia/metabolism , Nerve Fibers, Myelinated/metabolism , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/metabolism , Cells, Cultured , Chronic Disease , Cystatins/deficiency , Cystatins/metabolism , Demyelinating Autoimmune Diseases, CNS/genetics , Demyelinating Autoimmune Diseases, CNS/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Neurologic Mutants , Mice, Transgenic , Microglia/pathology , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/genetics , Recovery of Function/genetics
12.
Ann Neurol ; 68(3): 369-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20641064

ABSTRACT

OBJECTIVE: Clinical studies indicate that anti-CD20 B-cell depletion may be an effective multiple sclerosis (MS) therapy. We investigated mechanisms of anti-CD20-mediated immune modulation using 2 paradigms of experimental autoimmune encephalomyelitis (EAE). METHODS: Murine EAE was induced by recombinant myelin oligodendrocyte glycoprotein (rMOG), a model in which B cells are considered to contribute pathogenically, or MOG peptide (p)35-55, which does not require B cells. RESULTS: In EAE induced by rMOG, B cells became activated and, when serving as antigen-presenting cells (APCs), promoted differentiation of proinflammatory MOG-specific Th1 and Th17 cells. B-cell depletion prevented or reversed established rMOG-induced EAE, which was associated with less central nervous system (CNS) inflammation, elimination of meningeal B cells, and reduction of MOG-specific Th1 and Th17 cells. In contrast, in MOG p35-55-induced EAE, B cells did not become activated or efficiently polarize proinflammatory MOG-specific T cells, similar to naive B cells. In this setting, anti-CD20 treatment exacerbated EAE, and did not impede development of Th1 or Th17 cells. Irrespective of the EAE model used, B-cell depletion reduced the frequency of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), and increased the proinflammatory polarizing capacity of remaining myeloid APCs. INTERPRETATION: Our study highlights distinct roles for B cells in CNS autoimmunity. Clinical benefit from anti-CD20 treatment may relate to inhibition of proinflammatory B cell APC function. In certain clinical settings, however, elimination of unactivated B cells, which participate in regulation of T cells and other APC, may be undesirable. Differences in immune responses to MOG protein and peptide may be important considerations when choosing an EAE model for testing novel B cell-targeting agents for MS.


Subject(s)
Antibodies/therapeutic use , Antigens, CD20/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Lymphocyte Activation/immunology , Animals , Antigens, CD20/genetics , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Flow Cytometry/methods , Forkhead Transcription Factors/metabolism , Glycoproteins/adverse effects , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/adverse effects , Statistics, Nonparametric , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
13.
Neuroimage ; 49(2): 1180-9, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19796690

ABSTRACT

Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS). Despite progress in understanding immunogenetic aspects of this disease, the mechanisms involved in lesion formation are unknown. To gain new insights into the neuropathology of MS, we used an innovative integration of Fourier transform infrared (FT-IR) microspectroscopy, bioinformatics, and a synchrotron light source to analyze macromolecular changes in the CNS during the course and prevention of experimental autoimmune encephalomyelitis (EAE), an animal model for MS. We report that subtle chemical and structural changes not observed by conventional histology were detected before the onset of clinical signs of EAE. Moreover, trained artificial neural networks (ANNs) could discriminate, with excellent sensitivity and specificity, pathology from surrounding tissues and the early stage of the disease progression. Notably, we show that this novel measurement platform can detect characteristic differences in biochemical composition of lesion pathology in animals partially protected against EAE by vaccination with Nogo-A, an inhibitor of neural outgrowth, demonstrating the potential for automated screening and evaluation of new therapeutic agents.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Animals , Automation , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Image Processing, Computer-Assisted/methods , Mice , Mice, Inbred C57BL , Multiple Sclerosis , Myelin Proteins/immunology , Myelin Proteins/therapeutic use , Neural Networks, Computer , Nogo Proteins , Peptides/immunology , Peptides/therapeutic use , Sensitivity and Specificity , Spectroscopy, Fourier Transform Infrared/methods , Time Factors , Vaccination
14.
J Immunol ; 181(11): 7571-80, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017946

ABSTRACT

Autoimmune diseases are incurable. We have hypothesized that these diseases can be cured by the transplantation of bone marrow (BM) stem cells that have been genetically engineered to express self-Ag. Here we have tested this hypothesis in experimental autoimmune encephalomyelitis (EAE) induced by the self-Ag myelin oligodendrocyte glycoprotein (MOG). We show that, in mice, transplantation of BM genetically modified to express MOG prevented the induction and progression of EAE, and combined with antecedent corticosteroid treatment, induced long-term remission of established disease. Mice remained resistant to EAE development upon subsequent rechallenge with MOG. Transfer of BM from these mice rendered recipients resistant to EAE. Splenocytes from these mice failed to proliferate or produce IL-17, IFN-gamma, and GM-CSF in response to MOG(35-55) peptide stimulation and they failed to produce MOG autoantibody. Mechanistically, we demonstrated in vivo reduction in development of CD4(+) MOG(35-55)-specific thymocytes, indicative of clonal deletion with no evidence for selection of Ag-specific regulatory T cells. These findings validate our hypothesis that transplantation of genetically modified BM expressing disease-causative self-Ag provides a curative approach by clonal deletion of disease-causative self-reactive T cells.


Subject(s)
Autoantigens/immunology , Bone Marrow Transplantation , Clonal Deletion/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Glycoproteins/immunology , Immune Tolerance/immunology , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/immunology , Adrenal Cortex Hormones/pharmacology , Animals , Autoantibodies/genetics , Autoantibodies/immunology , Autoantigens/genetics , Clonal Deletion/drug effects , Clonal Deletion/genetics , Cytokines/genetics , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Gene Expression/genetics , Gene Expression/immunology , Glycoproteins/genetics , Immune Tolerance/drug effects , Immune Tolerance/genetics , Immunity, Innate/genetics , Immunity, Innate/immunology , Mice , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/genetics , Thymus Gland/immunology , Transduction, Genetic
15.
Nat Neurosci ; 7(7): 736-44, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15184901

ABSTRACT

Inhibitors associated with CNS myelin are thought to be important in the failure of axons to regenerate after spinal cord injury and in other neurodegenerative disorders. Here we show that targeting the CNS-specific inhibitor of neurite outgrowth Nogo A by active immunization blunts clinical signs, demyelination and axonal damage associated with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). Mice vaccinated against Nogo A produce Nogo-specific antibodies that block the neurite outgrowth inhibitory activity associated with CNS myelin in vitro. Passive immunization with anti-Nogo IgGs also suppresses EAE. Our results identify Nogo A as an important determinant of the development of EAE and suggest that its blockade may help to maintain and/or to restore the neuronal integrity of the CNS after autoimmune insult in diseases such as MS. Our finding that Nogo A is involved in CNS autoimmune demyelination indicates that this molecule may have a far more complex role than has been previously anticipated.


Subject(s)
Encephalomyelitis, Acute Disseminated , Myelin Proteins/physiology , Neurites/physiology , Animals , Animals, Newborn , Antibodies/therapeutic use , Cells, Cultured , Cytokines/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalomyelitis, Acute Disseminated/metabolism , Encephalomyelitis, Acute Disseminated/physiopathology , Encephalomyelitis, Acute Disseminated/therapy , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis , Myelin Proteins/genetics , Myelin Proteins/immunology , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Nogo Proteins , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Silver Staining/methods , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Time Factors , Vaccination/adverse effects
16.
Nat Biotechnol ; 21(9): 1033-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12910246

ABSTRACT

The diversity of autoimmune responses poses a formidable challenge to the development of antigen-specific tolerizing therapy. We developed 'myelin proteome' microarrays to profile the evolution of autoantibody responses in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS). Increased diversity of autoantibody responses in acute EAE predicted a more severe clinical course. Chronic EAE was associated with previously undescribed extensive intra- and intermolecular epitope spreading of autoreactive B-cell responses. Array analysis of autoantigens targeted in acute EAE was used to guide the choice of autoantigen cDNAs to be incorporated into expression plasmids so as to generate tolerizing vaccines. Tolerizing DNA vaccines encoding a greater number of array-determined myelin targets proved superior in treating established EAE and reduced epitope spreading of autoreactive B-cell responses. Proteomic monitoring of autoantibody responses provides a useful approach to monitor autoimmune disease and to develop and tailor disease- and patient-specific tolerizing DNA vaccines.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoassay/methods , Myelin Sheath/immunology , Protein Array Analysis/methods , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Animals , Drug Tolerance , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Mice , Multiple Sclerosis/diagnosis , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Protein Interaction Mapping/methods , Treatment Outcome
17.
Neurol Neuroimmunol Neuroinflamm ; 3(2): e212, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27027096

ABSTRACT

OBJECTIVE: To investigate the role of very late antigen-4 (VLA-4) on regulatory B cells (Breg) in CNS autoimmune disease. METHODS: Experimental autoimmune encephalomyelitis (EAE) was induced in mice selectively deficient for VLA-4 on B cells (CD19cre/α4(f/f)) by immunization with myelin oligodendrocyte glycoprotein (MOG) peptide (p)35-55 or recombinant human (rh) MOG protein. B-cell and T-cell populations were examined by flow cytometry and immunohistochemistry. Breg were evaluated by intracellular IL-10 staining of B cells and, secondly, by coexpression of CD1d and CD5. RESULTS: As previously reported, EAE was less severe in B-cell VLA-4-deficient vs control CD19cre mice when induced by rhMOG, a model that is B-cell-dependent and leads to efficient B-cell activation and antibody production. Paradoxically, B-cell VLA-4-deficient mice developed more severe clinical disease than control mice when EAE was induced with MOG p35-55, a B-cell-independent encephalitogen that does not efficiently activate B cells. Peripheral T-cell and humoral immune responses were not altered in B-cell VLA-4-deficient mice. In MOG p35-55-induced EAE, B-cell VLA-4 deficiency reduced CNS accumulation of B but not T cells. Breg were detected in the CNS of control mice with MOG p35-55-induced EAE. However, more severe EAE in B-cell VLA-4-deficient mice was associated with virtual absence of CNS Breg. CONCLUSIONS: Our results demonstrate that CNS accumulation of Breg is VLA-4-dependent and suggest that Breg may contribute to regulation of CNS autoimmunity in situ. These observations underscore the need to choose the appropriate encephalitogen when studying how B cells contribute to pathogenesis or regulation of CNS autoimmunity.

18.
Neurol Neuroimmunol Neuroinflamm ; 3(5): e272, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27704036

ABSTRACT

OBJECTIVE: To evaluate the influence of oral laquinimod, a candidate multiple sclerosis (MS) treatment, on induction of T follicular helper cells, development of meningeal B cell aggregates, and clinical disease in a spontaneous B cell-dependent MS model. METHODS: Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice by immunization with recombinant myelin oligodendrocyte glycoprotein (rMOG) protein. Spontaneous EAE was evaluated in C57BL/6 MOG p35-55-specific T cell receptor transgenic (2D2) × MOG-specific immunoglobulin (Ig)H-chain knock-in (IgHMOG-ki [Th]) mice. Laquinimod was administered orally. T cell and B cell populations were examined by flow cytometry and immunohistochemistry. RESULTS: Oral laquinimod treatment (1) reduced CD11c+CD4+ dendritic cells, (2) inhibited expansion of PD-1+CXCR5+BCL6+ T follicular helper and interleukin (IL)-21-producing activated CD4+CD44+ T cells, (3) suppressed B cell CD40 expression, (4) diminished formation of Fas+GL7+ germinal center B cells, and (5) inhibited development of MOG-specific IgG. Laquinimod treatment not only prevented rMOG-induced EAE, but also inhibited development of spontaneous EAE and the formation of meningeal B cell aggregates. Disability progression was prevented when laquinimod treatment was initiated after mice developed paralysis. Treatment of spontaneous EAE with laquinimod was also associated with increases in CD4+CD25hiFoxp3+ and CD4+CD25+IL-10+ regulatory T cells. CONCLUSIONS: Our observations that laquinimod modulates myelin antigen-specific B cell immune responses and suppresses both development of meningeal B cell aggregates and disability progression in spontaneous EAE should provide insight regarding the potential application of laquinimod to MS treatment. Results of this investigation demonstrate how the 2D2 × Th spontaneous EAE model can be used successfully for preclinical evaluation of a candidate MS treatment.

19.
Sci Rep ; 6: 34594, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708418

ABSTRACT

Studies in experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS), have shown that regulatory B cells modulate the course of the disease via the production of suppressive cytokines. While data indicate a role for transforming growth factor (TGF)-ß1 expression in regulatory B cell functions, this mechanism has not yet been tested in autoimmune neuroinflammation. Transgenic mice deficient for TGF-ß1 expression in B cells (B-TGF-ß1-/-) were tested in EAE induced by recombinant mouse myelin oligodendrocyte glycoprotein (rmMOG). In this model, B-TGF-ß1-/- mice showed an earlier onset of neurologic impairment compared to their littermate controls. Exacerbated EAE susceptibility in B-TGF-ß1-/- mice was associated with augmented CNS T helper (Th)1/17 responses. Moreover, selective B cell TGF-ß1-deficiency increased the frequencies and activation of myeloid dendritic cells, potent professional antigen-presenting cells (APCs), suggesting that B cell-derived TGF-ß1 can constrain Th1/17 responses through inhibition of APC activity. Collectively our data suggest that B cells can down-regulate the function of APCs, and in turn encephalitogenic Th1/17 responses, via TGF-ß1, findings that may be relevant to B cell-targeted therapies.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta1/genetics , Animals , B-Lymphocytes, Regulatory/pathology , Cell Communication/immunology , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Deletion , Gene Expression , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Th1 Cells/pathology , Th17 Cells/pathology , Transforming Growth Factor beta1/immunology
20.
J Neuroimmunol ; 162(1-2): 122-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15833367

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) models multiple sclerosis (MS) and is characterized by marked mononuclear cell influx in the brain. Several studies have demonstrated a role for chemokines during EAE. It remains to be determined whether these mediators modulate EAE primarily by mediating leukocyte influx into the CNS or by modifying lymphocyte activation and/or trafficking into lymphoid organs. After induction of EAE with MOG(35-55), leukocyte recruitment peaked on day 14 and correlated with symptom onset, TNF-alpha production and production of CCL2 and CCL5. Levels of CXCL-10 and CCL3 were not different from control animals. Using intravital microscopy, we demonstrated that leukocyte rolling and adhesion also peaked at day 14. Treatment with anti-CCL2 or anti-CCL5 antibodies just prior to the intravital microscopy prevented leukocyte adhesion, but not rolling. Our data suggest that induction of leukocyte adhesion to the brain microvasculature is an important mechanism by which CCL2 and CCL5 participate in the pathophysiology of EAE.


Subject(s)
Chemokine CCL2/metabolism , Chemokines, CC/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Leukocytes/metabolism , Analysis of Variance , Animals , Antibodies/pharmacology , Body Weight/drug effects , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CCL2/immunology , Chemokine CCL5/metabolism , Chemokines, CC/immunology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Enzyme-Linked Immunosorbent Assay/methods , Female , Glycoproteins/metabolism , Leukocytes/drug effects , Mice , Mice, Inbred C57BL , Microcirculation/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL