Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell Physiol Biochem ; 47(1): 390-402, 2018.
Article in English | MEDLINE | ID: mdl-29772575

ABSTRACT

BACKGROUND/AIMS: This study aimed to explore whether the adoptive transfusion of autologous CD4+CD25+ regulatory T cells (CD4+CD25+ Tregs) has a therapeutic effect on Experimental autoimmune neuritis (EAN) model rats, and it provides new experimental and theoretical bases for the immunotherapy of Guillain-Barre syndrome (GBS). METHODS: CD4+CD25+ Tregs were sorted from the spleens of rats using immunomagnetic bead separation techniques combined with flow cytometry. Their in vitro inhibitory function was determined using a lymphocyte proliferation inhibition test, and their purity was confirmed by flow cytometry. Cells were stimulated using CD3/CD28 monoclonal antibodies and were cultured in culture medium containing interleukin 2 (IL-2), transforming growth factor-ß (TGF-ß) and rapamycin. After 15 days of amplification, CD4+CD25+ Tregs were collected and transfused into EAN model rats. Changes in the pathology and electron microscopical morphology of rat sciatic nerves in the normal group, untreated group, low-dose group (2 × 107) and high-dose group (4 × 107) were observed, and the expression of CD4+CD25+FOXP3 in peripheral blood in the four groups of rats was detected by flow cytometry. RESULTS: Compared with rats in the untreated group, rats in the treatment groups had significantly reduced infiltration of inflammatory cells in the sciatic nerve, as well as myelin and axonal damage. Additionally, the CD4+CD25+ Tregs levels in peripheral blood were significantly higher than those in the untreated group (P< 0. 05). Moreover, the therapeutic effect became more significant with an increase in the dose of adoptive transfusion. CONCLUSION: Adoptive transfusion of CD4+CD25+ Tregs into EAN model rats has significant therapeutic effects.


Subject(s)
Adoptive Transfer , CD4 Antigens/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Neuritis, Autoimmune, Experimental/therapy , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Adoptive Transfer/methods , Animals , Cells, Cultured , Disease Models, Animal , Female , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew
2.
J Autoimmun ; 92: 114-125, 2018 08.
Article in English | MEDLINE | ID: mdl-29884340

ABSTRACT

Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune disease of the peripheral nerves evolving with diffuse sensory and motor symptoms. Although it is claimed that in neurodegenerative pathologies, a common feature is the failure of proteolytic systems to adequately eliminate aggregated or misfolded proteins, it has not been addressed whether autophagy, a central "clearance" system delivering damaged intracellular components to lysosomes, is affected in CIDP. The focus of the present investigation was therefore to determine if some defects exist in autophagy processes in this setting and if they can be corrected or minimized using an appropriate treatment targeting this survival pathway. Experiments were performed using a rat model mimicking human CIDP, also known as chronic experimental autoimmune neuritis (c-EAN), the disease establishment and development of which was followed at both the clinical and biological levels (indices of disease severity, histopathological alteration, cytokines and antibodies rates). Based on immunofluorescence and western immunoblotting experiments on sciatic nerves and spleen cells from c-EAN rats, we demonstrate that both, macroautophagy and chaperone-mediated autophagy (CMA), are significantly altered in non-neuronal cells of the peripheral nervous system. We show further that a 21-mer synthetic phosphopeptide called P140, known to target CMA and successfully used in pathological settings where CMA markers are overexpressed, considerably ameliorates the clinical and biological course of the disease in c-EAN rats. P140 displayed prophylactic and therapeutic effects, both in terms of disease intensity and chronicity, and preserved sciatic nerves from disease-related damages. Our findings uncover new disrupted molecular pathways in a c-EAN model and provide a proof-of-concept that targeting CMA might represent a promising therapeutic strategy for treating inflammatory neuropathies for which no disease-specific treatment is currently available.


Subject(s)
Biological Products/therapeutic use , Neuritis, Autoimmune, Experimental/therapy , Peptide Fragments/therapeutic use , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Sciatic Nerve/physiology , Animals , Autophagy , Disease Models, Animal , Disease Progression , Humans , Male , Motor Activity , Proteostasis Deficiencies , Rats , Rats, Inbred Lew , Sciatic Nerve/drug effects
3.
J Immunol ; 197(10): 3831-3840, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27798164

ABSTRACT

Programmed death 1 (PD-1; CD279), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for T cell dysfunction in infectious diseases and cancers. The ligand for PD-1, programmed death ligand 1 (PD-L1; also known as B7-H1, CD274), is a member of the B7 family. The engagement of PD-1 with programmed death ligand can downregulate autoreactive T cells that participate in multiple autoimmune diseases. Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome, and the pathogenesis of EAN is mediated principally through T cells and macrophages. In this study, we investigated the effects of PD-L1 in EAN rats. For preventative and therapeutic management, we administered PD-L1, which successfully decreased the severity of EAN; it alleviated the neurologic course of EAN, as well as inhibited the infiltration of inflammatory cells and demyelination of sciatic nerves. Our data revealed that PD-L1 treatment inhibited lymphocyte proliferation and altered T cell differentiation by inducing decreases in IFN-γ+CD4+ Th1 cells and IL-17+CD4+ Th17 cells and increases in IL-4+CD4+ Th2 cells and Foxp3+CD4+ regulatory T cells. The expression levels of p-STAT3 and Foxp3 were significantly different in PD-L1-treated groups compared with the control group. Additionally, PD-L1 regulated the expression of Foxp3 and p-STAT3 in EAN, probably by inhibiting PI3K/AKT/mTOR signaling expression. In summary, PD-L1 is a potentially useful agent for the treatment of EAN because of its anti-inflammatory and neuroprotective effects.


Subject(s)
B7-H1 Antigen/pharmacology , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/therapy , Peripheral Nervous System/immunology , Animals , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Demyelinating Diseases/prevention & control , Disease Models, Animal , Gene Expression Regulation , Guillain-Barre Syndrome/immunology , Interferon-gamma/drug effects , Interleukin-17/immunology , Interleukin-4/immunology , Lymphocyte Activation , Neuritis, Autoimmune, Experimental/physiopathology , Rats , Sciatic Nerve/drug effects , T-Lymphocytes, Regulatory , Th17 Cells/drug effects , Th17 Cells/immunology , Th2 Cells
4.
Clin Exp Immunol ; 175(2): 181-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24116957

ABSTRACT

Spontaneous autoimmune polyneuropathy (SAP) in B7-2 knock-out non-obese diabetic (NOD) mice is mediated by myelin protein zero (P0)-reactive T helper type 1 (Th1) cells. In this study, we investigated the role of B cells in SAP, focusing on CD19 as a potential therapeutic target. We found that P0-specific plasmablasts and B cells were increased in spleens of SAP mice compared to wild-type NOD mice. Depletion of B cells and plasmablasts with anti-CD19 monoclonal antibody (mAb) led to attenuation of disease severity when administered at 5 months of age. This was accompanied by decreased serum immunoglobulin (Ig)G and IgM levels, depletion of P0-specific plasmablasts and B cells, down-regulation/internalization of surface CD19 and increased frequency of CD4(+) regulatory T cells in spleens. We conclude that B cells are crucial to the pathogenesis of SAP, and that CD19 is a promising B cell target for the development of disease-modifying agents in autoimmune neuropathies.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes/immunology , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/therapy , Polyneuropathies/immunology , Polyneuropathies/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity/immunology , Antigens, CD19/metabolism , Cell Proliferation , Down-Regulation , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Mice, Inbred NOD , Mice, Knockout , Myelin P0 Protein/immunology , Plasma Cells/immunology , Th1 Cells/immunology
5.
Immunol Invest ; 41(2): 171-82, 2012.
Article in English | MEDLINE | ID: mdl-21864114

ABSTRACT

Susceptible-strain animals immunized with P2 peptide could generate the disease of experimental autoimmune neuritis (EAN) with inflammation and demyelination of peripheral nerve. A myriad of transcription factors and inflammatory cytokines have been found to participate in this process; however, the roles of toll-like receptors (TLRs) are poorly understood in EAN. The aim of this study is to explore the role of TLR9 in the pathogenesis of EAN. The EAN was induced in Lewis rat by immunization with P2(53-78) and complete Freund's adjuvant. CpG oligodeoxynucleotides (ODN) (cODN), a suppressive ODN (sODN) and a control non-specific ODN (nODN) were respectively administered to explore the role of TLR9 in EAN both in vivo and vitro. Following immunization up to the peak phase of EAN, EAN rats inoculated with sODN had remarkably better clinical score of EAN and expressed a significantly inhibited TLR9 signaling pathway. Our study suggests that TLR9 may be involved in the pathogenesis of EAN.


Subject(s)
Guillain-Barre Syndrome/immunology , Neuritis, Autoimmune, Experimental/immunology , Oligodeoxyribonucleotides, Antisense/administration & dosage , Toll-Like Receptor 9/metabolism , Animals , Disease Models, Animal , Disease Progression , Disease Susceptibility , Freund's Adjuvant/administration & dosage , Guillain-Barre Syndrome/therapy , Humans , Immunization , Male , Myelin P2 Protein/administration & dosage , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/physiopathology , Neuritis, Autoimmune, Experimental/therapy , Oligodeoxyribonucleotides, Antisense/adverse effects , Peptide Fragments/administration & dosage , Rats , Rats, Inbred Lew , Toll-Like Receptor 9/genetics
6.
J Biomed Biotechnol ; 2011: 294046, 2011.
Article in English | MEDLINE | ID: mdl-20981263

ABSTRACT

Optic neuritis associated with multiple sclerosis has a strong association with organ-specific autoimmune disease. The goal of our research is to establish an optimal organ-specific animal model to elucidate the pathogenetic mechanisms of the disease and to develop therapeutic strategies using the model. This paper is divided into five sections: (1) clinical picture of optic neuritis associated with multiple sclerosis, (2) elucidation of pathogenesis using animal models with inflammation in optic nerve and spinal cord, (3) clinical relevance of concurrent encephalomyelitis in optic neuritis model, (4) retinal damage in a concurrent multiple sclerosis and optic neuritis model, and (5) development of novel therapies using mouse optic neuritis model. Advanced therapies using biologicals have succeeded to control intractable optic neuritis in animal models. This may ultimately lead to prevention of vision loss within a short period from acute onset of optic neuritis in human. By conducting research flexibly, ready to switch from the bench to the bedside and from the bedside to the bench as the opportunity arises, this strategy may help to guide the research of optic neuritis in the right direction.


Subject(s)
Neuritis, Autoimmune, Experimental/etiology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Inflammation/complications , Inflammation/pathology , Mice , Multiple Sclerosis/complications , Multiple Sclerosis/pathology , Neuritis, Autoimmune, Experimental/therapy , Optic Nerve/pathology , Retina/pathology , Spinal Cord/pathology
7.
Nat Med ; 1(11): 1162-6, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7584988

ABSTRACT

Nerve-specific autoimmune T lymphocytes were used as vehicles to deliver therapeutically useful neurotrophic factors across the endothelial blood-nerve barrier. P2 protein-reactive T-lymphocyte lines from Lewis rats were transduced with a recombinant retrovirus containing the mouse nerve growth factor (NGF) gene. The engineered T cells released high amounts of NGF dependent on antigenic stimulation in vitro. After intravenous injection, the T cells infiltrated the rat peripheral nervous system and persisted there for at least two weeks. Local release of NGF from engineered T cells was demonstrable by immunocytochemistry and by an anti-inflammatory effect on infiltrating macrophages.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Transfer Techniques , Genetic Therapy/methods , Myelin P2 Protein/immunology , Nerve Growth Factors/genetics , Neuritis, Autoimmune, Experimental/therapy , Animals , Capillary Permeability , Cell Movement , Immunoassay , Immunohistochemistry , In Situ Hybridization , Macrophages/immunology , Mice , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/pharmacology , Neuritis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew , Sciatic Nerve/pathology
8.
J Mol Neurosci ; 71(4): 713-723, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32915416

ABSTRACT

Nerve demyelination or axonal lesions are characteristic of experimental autoimmune neuritis (EAN). Previous studies have demonstrated that microRNA-338 can regulate the differentiation and maturation of oligodendrocytes and Schwann cells and promote injured peripheral nerves in rats. In this study, we used microRNA-338 coded lentivirus vector (miR-338-LV) in a Lewis rat EAN model, in with the conjunction P0 peptide 180-199 which was injected into the footpads of animals to induce immunization. The clinical scores of miR-338-LV and intravenous immunoglobulin (IVIg) (positive drug) groups were significantly superior to those of untreated group at disease peak and disease plateau (p < 0.05). The nerve conduction velocity and the compound nerve action potential amplitude of miR-338-LV and IVIg groups increased significantly compared to those of the untreated group at disease peak (p < 0.01). At disease peak, myelin swelling, cavity formation, and lamellae separation showed improvement in miR-338-LV and IVIg groups compared to untreated group. S100 and NF200 expression in miR-338-LV and IVIg groups increased compared to that in untreated group. Iba1 and S100 co-expression in Schwann cells in miR-338-LV and IVIg groups decreased compared to that in untreated group, which was indicative of the reduced conversion of Schwann cells into inflammatory cells. Overall, miR-338-LV in sciatic nerves might improve neuromuscular function in EAN by inhibiting the conversion of Schwann cells into inflammatory cells.


Subject(s)
MicroRNAs/genetics , Neuritis, Autoimmune, Experimental/therapy , Sciatic Nerve/metabolism , Animals , Calcium-Binding Proteins/metabolism , Female , MicroRNAs/metabolism , Microfilament Proteins/metabolism , Myelin Sheath/metabolism , Nerve Regeneration , RNAi Therapeutics/methods , Rats , Rats, Inbred Lew , S100 Proteins/metabolism , Schwann Cells/metabolism , Sciatic Nerve/physiology , Transfection/methods
9.
J Neurosci Res ; 88(8): 1651-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20077429

ABSTRACT

Campylobacteriosis is a frequent antecedent event in Guillain-Barré syndrome (GBS), inducing high-titer serum antibodies for ganglioside antigens in the peripheral nervous system (PNS). Molecular mimicry between the lipooligosaccharide (LOS) component of Campylobacter jejuni and human peripheral nerve gangliosides is believed to play an important role in the pathogenesis of GBS. Conventional treatment strategies for patients with GBS include plasmapheresis, intravenous immunoglobulin (IVIG), and immunosuppression, which are invasive or relatively ineffective. In this study, we used our animal model of GBS, in which Lewis rats were immunized with GD3-like LOS isolated from C.jejuni. The animals developed anti-GD3 ganglioside antibodies and manifested neuromuscular dysfunction. To develop novel therapeutic strategies, we treated the animals by intraperitoneal administration of an anti-GD3 antiidiotype monoclonal antibody (BEC2) that specifically interacts with the pathogenic antibody. The treated animals had a remarkable reduction of anti-GD3 antibody titers and improvement of motor nerve functions. The results suggest that ganglioside mimics, such as antiidiotype antibodies, may be powerful reagents for therapeutic intervention in GBS by neutralizing specific pathogenic antiganglioside antibodies.


Subject(s)
Antibodies, Anti-Idiotypic/therapeutic use , Gangliosides/immunology , Neuritis, Autoimmune, Experimental/therapy , Action Potentials/drug effects , Action Potentials/physiology , Animals , Biotinylation/methods , Campylobacter jejuni/immunology , Coculture Techniques/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Ether-A-Go-Go Potassium Channels/immunology , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/pharmacokinetics , Female , Freund's Adjuvant/immunology , Lipopolysaccharides , Motor Neurons/pathology , Motor Neurons/ultrastructure , Muscle, Skeletal/physiology , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacokinetics , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/complications , Neuritis, Autoimmune, Experimental/immunology , Neuromuscular Junction Diseases/drug therapy , Neuromuscular Junction Diseases/etiology , Neuromuscular Junction Diseases/metabolism , Organ Culture Techniques , Rats , Rotarod Performance Test/methods , Sciatic Nerve/pathology , Sciatic Nerve/ultrastructure , Spinal Cord/physiology , Time Factors
10.
Int Immunopharmacol ; 61: 290-296, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29908492

ABSTRACT

BACKGROUND: Guillain-Barré syndrome (GBS) is a rare, autoimmune-mediated disease. The use of Bifidobacterium is reportedly effective in alleviating GBS since they act by regulating T helper (Th) cells. OBJECTIVES: In this study, we explored the differentiation of T helper cell subsets in patients with GBS. We also evaluated the effect of GBS on Bifidobacterium levels in patients and the likely protective influence of this bacterium in alleviating the disease in an animal model. MATERIALS AND METHODS: We used flow cytometry, and real-time polymerase chain reaction (PCR) to determine the T cell subsets differentiation among 30 GBS patients and 20 healthy controls (HC). The concentration of Bifidobacterium was assayed by real-time PCR. Experimental autoimmune neuritis (EAN) animal model was established to support the protective role of Bifidobacterium in GBS. RESULTS: The expression of Th cells, Th2 and Th17 in the patients was significantly higher than that in the HC, while Treg cells decreased substantially. Moreover, the levels of Bifidobacterium in the GBS patients were considerably lower than those in the HC, the concentration of Bifidobacterium correlating with Th2 and Th17 subsets negatively. Treatment with Bifidobacterium significantly reduced the levels of Th2 and Th17 and promoted the levels of Treg cells. CONCLUSIONS: We concluded from this study that Bifidobacterium alleviated GBS by regulating Th cells, although in-depth studies might be required to fully understand the mechanism of action.


Subject(s)
Bifidobacterium longum subspecies infantis/immunology , Biological Therapy , Guillain-Barre Syndrome/therapy , Neuritis, Autoimmune, Experimental/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Animals , Autoimmunity , Cell Differentiation , Cells, Cultured , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Rats , Rats, Inbred Strains , T-Lymphocyte Subsets/microbiology , T-Lymphocytes, Regulatory/microbiology
11.
Expert Opin Ther Targets ; 22(7): 567-577, 2018 07.
Article in English | MEDLINE | ID: mdl-29856236

ABSTRACT

INTRODUCTION: Macrophage migration inhibitory factor (MIF) plays an important role in the pathogenesis of Guillain-Barré syndrome (GBS) and its animal model experimental autoimmune neuritis (EAN), which may offer an opportunity for the development of the novel therapeutic strategies for GBS. Areas covered: 'macrophage migration inhibitory factor' and 'Guillain-Barré syndrome' were used as keywords to search for related publications on Pub-Med, National Center for Biotechnology Information (NCBI), USA. MIF is involved in the etiology of various inflammatory and autoimmune disorders. However, the roles of MIF in GBS and EAN have not been summarized in the publications we identified. Therefore, in this review, we described and analyzed the major roles of MIF in GBS/EAN. Primarily, this molecule aggravates the inflammatory responses in this disorder. However, multiple studies indicated a protective role of MIF in GBS. The potential of MIF as a therapeutic target in GBS has been recently demonstrated in experimental and clinical studies, although clinical trials have been unavailable to date. Expert opinion: MIF plays a critical role in the initiation and progression of GBS and EAN, and it may represent a potential therapeutic target for GBS.


Subject(s)
Guillain-Barre Syndrome/physiopathology , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Neuritis, Autoimmune, Experimental/physiopathology , Animals , Disease Models, Animal , Disease Progression , Drug Development/methods , Guillain-Barre Syndrome/therapy , Humans , Neuritis, Autoimmune, Experimental/therapy
12.
J Neuroimmunol ; 168(1-2): 168-74, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16171874

ABSTRACT

Macrophage migration inhibitory factor (MIF) is a proinflammatory type 1 cytokine that plays a pathogenic role in several inflammatory and autoimmune diseases. The role of this cytokine in peripheral nerve inflammatory disease has not been evaluated. Therefore, to evaluate the role of macrophage migration inhibitory factor (MIF) in Guillain-Barré syndrome (GBS) and experimental allergic neuritis (EAN), we determined MIF circulating levels in a series of patients with GBS and healthy subjects with ELISA and evaluated the effect of two specific inhibitors of MIF, a neutralizing monoclonal antibody or a chemical inhibitor ISO1 on the course of murine EAN. The data show increased MIF plasma levels in GBS patients as compared to healthy controls (p<0.0001) and a progressive increase of MIF circulating concentration with patient's disability (p<0.0001). Both anti-MIF mAb and ISO1 favorably influenced the course of EAN. Treated mice had a lower cumulative severity score (p=0.001) and reduced disease duration than the control mice (p<0.03). MIF may promote immune reaction in GBS. Therapeutic effects of both anti-MIF mAb and ISO1 in EAN suggest that MIF could be a promising therapeutic target in inflammatory demyelinating peripheral nerve disorders.


Subject(s)
Guillain-Barre Syndrome/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Neuritis, Autoimmune, Experimental/metabolism , Analysis of Variance , Animals , Antibodies, Monoclonal/therapeutic use , Disability Evaluation , Enzyme-Linked Immunosorbent Assay/methods , Guillain-Barre Syndrome/therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/immunology , Mice , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/therapy , Prospective Studies , Time Factors
13.
Immunol Lett ; 166(1): 13-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25952624

ABSTRACT

Toll-like receptor (TLR) 9, recognizing different ligands, confers distinct features of plasmacytoid dendritic cells (pDCs). Our previous study demonstrated a role for TLR9 in the mechanism of experimental autoimmune neuritis (EAN). In this study, we explored whether suppressive oligodeoxynucleotides (sODN) could induce tolerogenic pDCs via TLR9 and thus promote the recovery of EAN. Effects of different TLR9 ligands, CpG ODN and sODN on P0 180-199 peptide-stimulated pDCs were measured by detecting the expression of co-stimulatory molecules, indoleamine 2,3-dioxygenase (IDO), secretion of Th1- and Th2-type cytokines and the TLR9 signaling pathway. CpG ODN- or sODN-treated pDCs were intravenously injected into the EAN mice and their effects were compared. Our data showed that P0180-199 peptides significantly promoted mRNA expression of co-stimulatory molecules (CD40, CD80 and CD86) in pDCs and induced secretion of Th1-type cytokines. Treatment of CpG ODN aggravated the effects of P0 180-199 peptides on pDCs; however, sODN had the opposite effects and significantly upregulated the IDO expression in pDCs. Further analysis showed that MYD88 is necessary for sODN to modulate the TLR9/NF-κB signaling in pDCs. Finally, the sODN-treated pDCs significantly promoted recovery of the EAN mice. Taken together, sODN could induce tolerogenic pDCs and thus ameliorate the EAN.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/immunology , Neuritis, Autoimmune, Experimental/immunology , Oligodeoxyribonucleotides/pharmacology , Peptides/pharmacology , Animals , B7-1 Antigen/genetics , B7-2 Antigen/genetics , CD40 Antigens/genetics , Cells, Cultured , CpG Islands/genetics , Cytokines/biosynthesis , Cytokines/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Neuritis, Autoimmune, Experimental/therapy , RNA, Messenger/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Toll-Like Receptor 9/immunology , Transcription Factor RelA/metabolism
14.
Immunol Res ; 17(1-2): 217-27, 1998.
Article in English | MEDLINE | ID: mdl-9479583

ABSTRACT

Animal models of autoimmune diseases have greatly improved our current understanding of the pathogenesis of human autoimmunity and have provided the potential for therapies based on manipulation of the immune system. In our laboratory, we have investigated the immunopathogenesis of autoimmune diseases of the nervous system and muscle. We have developed immune-based approaches for the suppression of experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS), and experimental autoimmune neuritis (EAN), a model for the Guillain-Barré syndrome (GBS). These approaches included induction of peripheral tolerance, immunotoxin targeting of activated T cells, and cytokine manipulations. In addition, we identified the antigen and characterized immunopathologically an autoimmune inflammatory disease of skeletal muscle, experimental autoimmune myositis (EAM), a model for the human inflammatory muscle disease polymyositis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunotherapy , Multiple Sclerosis/immunology , Neuritis, Autoimmune, Experimental/immunology , Polyradiculoneuropathy/immunology , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/therapy , Humans , Mice , Multiple Sclerosis/therapy , Neuritis, Autoimmune, Experimental/therapy , Polymyositis/immunology , Polyradiculoneuropathy/therapy
15.
J Neuroimmunol ; 76(1-2): 112-6, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9184640

ABSTRACT

Experimental autoimmune neuritis (EAN) is an animal model for Guillain-Barré syndrome (GBS). Intravenous immunoglobulins (IVIg) are an effective treatment for GBS, but their mechanism of action is not well understood. Here we tested whether IVIg treatment, a potent modulator of proinflammatory assaults, reduces inflammation in EAN. The evaluation of IVIg treatment failed to demonstrate a salutary effect in different models of EAN. IVIg appears not to suppress the acute inflammatory insult on the peripheral nerve, but may have beneficial long-term effects not looked for in the present investigation.


Subject(s)
Immunoglobulins, Intravenous/therapeutic use , Neuritis, Autoimmune, Experimental/therapy , Animals , Female , Polyradiculoneuropathy/therapy , Rats , Rats, Inbred Lew
16.
J Neuroimmunol ; 78(1-2): 127-31, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9307236

ABSTRACT

To clarify how intravenous immunoglobulin (IVIg) acts on Guillain-Barré syndrome, we investigated the effects of intact-type IVIg treatment on experimental allergic neuritis (EAN) induced by immunizing with synthetic peptide from bovine P2 protein. Treatment with intact-type IVIg (400 mg/kg/day) on days 0, 7, 14, 15 and 16 after immunization prevented the paralysis, whereas treatment with F(ab')2 failed to alter the clinical course. Intact-type IVIg treatment given on days 0 and 1 showed almost the same efficacy. These results suggest that intact-type IVIg is superior to F(ab')2 in ameliorating the clinical course of EAN and that the Fc portion might affect the immune system.


Subject(s)
Immunoglobulin Fc Fragments/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Neuritis, Autoimmune, Experimental/therapy , Animals , Antibodies/analysis , Cattle , Immunization , Immunoglobulin Fc Fragments/immunology , Immunoglobulins, Intravenous/immunology , Male , Myelin P2 Protein/immunology , Neuritis, Autoimmune, Experimental/immunology , Peptide Fragments/immunology , Rats , Rats, Inbred Lew
17.
J Neuroimmunol ; 51(1): 69-75, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7512578

ABSTRACT

Experimental autoimmune neuritis (EAN) is a T cell-mediated autoimmune inflammatory disease of the peripheral nervous system that is characterized by demyelination and mononuclear cell infiltration. It is induced in Lewis rats by administration of myelin P2 protein or a synthetic peptide (SP-26) corresponding to amino acid residues 53-78 of bovine P2 protein. Recently, we showed that SP-26, when coupled to syngeneic spleen cells and administered intravenously, provided an effective means of inducing tolerance by inhibiting the clinical signs, decreased proliferative response of lymphoid cells to SP-26 and histological changes of EAN. However, our current data indicate that, despite tolerance induction in these Lewis rats, the antigen-specific delayed-type hypersensitivity (DTH) response to SP-26 remained intact. Furthermore, interferon (IFN)-gamma production by spleen cells of tolerized rats were unchanged as compared to EAN rats. The in vitro proliferation of T lymphocytes from tolerized rats stimulated by SP-26 was reduced as compared to EAN controls but was enhanced upon addition of exogenous interleukin-2. Thus, reduction in EAN clinical signs does not necessarily indicate a decrease in DTH response and IFN-gamma production in EAN Lewis rats. The implication of this finding in regard to immunoregulatory mechanism of DTH response is discussed.


Subject(s)
Cell Transplantation , Hypersensitivity, Delayed/therapy , Myelin Basic Protein/metabolism , Neuritis, Autoimmune, Experimental/therapy , Peptide Fragments/metabolism , Spleen/metabolism , Animals , Female , Hypersensitivity, Delayed/immunology , Immune Tolerance , Injections, Intravenous , Interferon-gamma/biosynthesis , Lymphocyte Activation , Myelin P2 Protein , Rats , Rats, Inbred Lew , Spleen/cytology , Transforming Growth Factor beta/pharmacology
18.
J Neuroimmunol ; 79(2): 129-37, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9394785

ABSTRACT

Experimental autoimmune neuritis (EAN) in Lewis rats is a T cell-mediated disease and serves as an animal model of human inflammatory demyelinating neuropathies. EAN can be induced by immunization with complete bovine peripheral nerve myelin (BPM), the myelin protein P2 or its neuritogenic peptide, each emulsified in complete Freund's adjuvant (CFA). The present study evaluates the effect of oral tolerization with BPM or P2 protein on the development of actively induced EAN. Oral administration of BPM strongly suppressed clinical and histological signs of EAN subsequently induced by BPM/CFA, but feeding of P2 protein alone did not affect its course. In contrast, feeding of BPM did not mitigate the course of EAN subsequently induced by immunization with neuritogenic P2 peptide/CFA. Oral therapy with BPM after onset of myelin-induced EAN only slightly ameliorated the further course of disease, but significantly reduced lethality of this severe form of disease. The findings suggest that immunogenicity of the antigens fed determine strength of tolerance, that downregulation of EAN occurs at the site of immunization and not in the nerve, and that active suppression rather than specific anergization is operative in mediating resistance to EAN. However, only partial tolerance to myelin-induced EAN was achieved in naive animals by transfer of spleen/LN cells from rats orally tolerized with BPM. Although methodic factors may have limited the effect of the cells, the result is suggestive of some contribution of anergy to oral tolerance in the present model. Cholera toxin and LPS were identified as oral adjuvants for BPM and prolonged the state of tolerance. However, LPS exhibited proinflammatory properties if EAN was induced early after BPM/LPS-feeding. Thus, oral application of a mixture of myelin components in combination with cholera toxin may be a useful treatment for chronic inflammatory neuropathies considered autoimmune in nature.


Subject(s)
Antigens/therapeutic use , Myelin Sheath/immunology , Neuritis, Autoimmune, Experimental/therapy , Adjuvants, Immunologic/pharmacology , Administration, Oral , Adoptive Transfer , Animals , Cattle , Cholera Toxin/therapeutic use , Drug Combinations , Female , Immune Tolerance/drug effects , Male , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/prevention & control , Peripheral Nerves/immunology , Rats , Rats, Inbred Lew
19.
J Neurol Sci ; 61(2): 149-60, 1983 Oct.
Article in English | MEDLINE | ID: mdl-6644324

ABSTRACT

Experimental allergic neuritis (EAN) is a demyelinating disease of the peripheral nervous system that can be induced in laboratory animals. This disorder has been considered to show many similarities to acute inflammatory polyneuropathy (Guillain-Barré syndrome, GBS). Reports that plasma exchange may benefit patients with GBS prompted the investigation of the effect of plasma exchange in EAN. A controlled study was performed on New Zealand White rabbits. Sixteen animals were allocated to control or treatment groups at the onset of the disease. Clinical assessment on days 7 and 14 showed that treated animals were less severely affected neurologically (P = 0.05, day 7; P less than 0.001 day 14), with a commensurate reduction in the severity of the histological lesions in peripheral nerves.


Subject(s)
Neuritis, Autoimmune, Experimental/therapy , Plasma Exchange , Animals , Female , Ganglia, Spinal/pathology , Nerve Fibers, Myelinated , Neuritis, Autoimmune, Experimental/pathology , Peripheral Nerves/pathology , Rabbits , Spinal Nerve Roots/pathology , Time Factors
20.
Rev Neurol (Paris) ; 155(10): 849-56, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10546299

ABSTRACT

It is now 83 years that Guillain, Barré and Strohl described the first two cases of an acute paralytic illness with the typical "dissociation albumino-cytologique" in the cerebrospinal fluid. Since then, the full spectrum of GBS has been documented in hundreds of cases ranging from acute inflammatory demyelinating polyneuropathy to the pure motor variants and the Miller Fisher syndrome. During the last 10 years, detailed immunopathologic features have been described and new triggering and possibly causative agents identified, the most prominent being the enteritic bacterium Campylobacter jejuni. Besides the pathogenic role of cell-mediated immunity, IgG antibodies have now been discovered which block neuromuscular transmission. Established treatments include plasma exchange therapy and intravenous immunoglobulin G. Together with sophisticated intensive care measures, mortality has now been reduced to below 5p. 100 with these treatment modalities. Several treatment strategies that proved effective in the animal model of GBS need to be studied in future clinical trials.


Subject(s)
Autoimmune Diseases/history , Guillain-Barre Syndrome/history , Animals , Antibody Formation , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/therapy , Clinical Trials as Topic , Forecasting , France , Glycolipids/immunology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/pathology , Guillain-Barre Syndrome/therapy , History, 20th Century , Humans , Immunity, Cellular , Immunoglobulins, Intravenous/therapeutic use , Models, Immunological , Models, Neurological , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/therapy , Plasma Exchange , Rats , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL