Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 79: 106051, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31863923

ABSTRACT

OBJECTIVE: Statins have immunomodulatory potential in autoimmune diseases but had not been studied as a disease-modifying agent in inflammatory myopathies. The objective of this study is to assess the effect of simvastatin in an experimental model of autoimmune myositis in mice on muscle strength and histopathology. METHODS: Four groups of mice (n = 5 per group) were selected for experimentally induced myositis. Mice were immunized with 1.5 mg myosin in complete Freund's adjuvant weekly for two times and injected with 500 ng pertussis toxin twice immediately after each immunization. From day 1 before immunization to 10 days after the last immunization, mice were treated with oral simvastatin (10 or 20 or 40 mg/kg) diluted in DMSO. The control group mice were injected with complete Freund's adjuvant weekly for two times and did not receive treatment. Non-immunized mice (n = 5 per group) were treated either with simvastatin (5 mg/kg or 20 mg/kg or 40 mg/kg of simvastatin diluted in DMSO) or with DMSO. RESULTS: Inflammation was observed in myositis groups with positive myositis-specific antibodies. Muscle strength dropped significantly after immunization. Immunized simvastatin 20 mg/kg treated group had significantly higher muscle strength versus non-treated myositis mice and versus other simvastatin doses. Besides, a trend toward higher serum Th17 percentage population was found in immunized non-treated mice, versus immunized simvastatin- treated mice, without significant difference. CONCLUSION: Simvastatin at 20 mg/kg decreases the severity of myositis in experimental autoimmune myositis and is a candidate of being a disease-modifying agent in inflammatory myopathies.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Nervous System Autoimmune Disease, Experimental/drug therapy , Simvastatin/therapeutic use , Th17 Cells/immunology , Animals , Autoantibodies/blood , Autoantigens/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Muscle Strength/drug effects , Myosins/immunology
2.
J Neuroimmune Pharmacol ; 14(4): 709-719, 2019 12.
Article in English | MEDLINE | ID: mdl-31435856

ABSTRACT

To explore the B cell depleting capacity of a low-dose (20 µg) subcutaneous mouse anti-CD20 antibody treatment on disease-relevant B cell populations within lymph nodes and the spleen. B cell depleting capacity was explored in healthy female C57BL/6 and BALB/c mice; following immune activation in two different mouse models: trinitrophenylated lipopolysaccharide model (thymus-independent response) and dinitrophenyl-keyhole limpet hemocyanin model (thymus-dependent response); and in a chronic neuroinflammation experimental autoimmune encephalomyelitis model. CD20 protein expression on B cell subpopulations was also studied. The subcutaneous anti-CD20 regimen resulted in rapid depletion of B cells in blood, lymph nodes and spleen. Low-dose subcutaneous treatment did not reduce antigen-specific immunoglobulin M and immunoglobulin G titers in all subgroups, and relatively spared splenic marginal zone (MZ) B cells in both T cell dependent and T cell independent B cell immunization models. Analysis of immune compartments during anti-CD20-modulated autoimmune neuroinflammation showed that the maximal B cell depletion was achieved within 2 days of treatment and was highest in the lymph node. Regardless of the tissues analyzed, low-dose subcutaneous treatment was characterized by rapid B cell repletion following treatment cessation. CD20 protein expression was consistent on all B cell subsets in blood, and was more pronounced in germinal center B cells of lymph nodes and MZ B-cells of the spleen. Low-dose subcutaneous anti-CD20 therapy effectively depleted B cells within lymphatic tissues and reduced the severity of neuroinflammation. These data suggest that subcutaneous anti-CD20 therapies can effectively target disease-relevant B cell populations, have shorter repletion kinetics and maintain vaccination responses, thereby achieving autoimmune amelioration without severely impacting immune surveillance functions. Graphical Abstract *p < 0.05; **p < 0.01. CD, cluster of differentiation; DNP-KLH, dinitrophenyl-keyhole limpet hemocyanin; EC50, concentration of a drug that gives half-maximal response; Ig, immunoglobulin; MZ, marginal zone; s.c., subcutaneous; SEM, standard error of mean; TNP-LPS, trinitrophenylatedlipopolysaccharide.


Subject(s)
Antigens, CD20/immunology , B-Lymphocyte Subsets/immunology , Nervous System Autoimmune Disease, Experimental/immunology , Animals , Antigens, CD20/metabolism , B-Lymphocyte Subsets/drug effects , Female , Hemocyanins/administration & dosage , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Injections, Subcutaneous , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nervous System Autoimmune Disease, Experimental/chemically induced , Nervous System Autoimmune Disease, Experimental/drug therapy , Treatment Outcome
3.
Acta Neuropathol ; 134(4): 655-666, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28623559

ABSTRACT

Dermatomyositis (DM) is an autoimmune disease associated with enhanced type I interferon (IFN) signalling in skeletal muscle, but the mechanisms underlying muscle dysfunction and inflammation perpetuation remain unknown. Transcriptomic analysis of early untreated DM muscles revealed that the main cluster of down-regulated genes was mitochondria-related. Histochemical, electron microscopy, and in situ oxygraphy analysis showed mitochondrial abnormalities, including increased reactive oxygen species (ROS) production and decreased respiration, which was correlated with low exercise capacities and a type I IFN signature. Moreover, IFN-ß induced ROS production in human myotubes was found to contribute to mitochondrial malfunctions. Importantly, the ROS scavenger N-acetyl cysteine (NAC) prevented mitochondrial dysfunctions, type I IFN-stimulated transcript levels, inflammatory cell infiltrate, and muscle weakness in an experimental autoimmune myositis mouse model. Thus, these data highlight a central role of mitochondria and ROS in DM. Mitochondrial dysfunctions, mediated by IFN-ß induced-ROS, contribute to poor exercise capacity. In addition, mitochondrial dysfunctions increase ROS production that drive type I IFN-inducible gene expression and muscle inflammation, and may thus self-sustain the disease. Given that current DM treatments only induce partial recovery and expose to serious adverse events (including muscular toxicity), protecting mitochondria from dysfunctions may open new therapeutic avenues for DM.


Subject(s)
Dermatomyositis/metabolism , Inflammation/metabolism , Interferon-beta/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adult , Aged , Animals , Cell Line , Cytokines/blood , Dermatomyositis/drug therapy , Dermatomyositis/pathology , Female , Free Radical Scavengers/pharmacology , Freund's Adjuvant , Humans , Inflammation/drug therapy , Inflammation/pathology , Male , Mice, Inbred BALB C , Middle Aged , Mitochondria/drug effects , Mitochondria/pathology , Muscle Weakness/drug therapy , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Nervous System Autoimmune Disease, Experimental/drug therapy , Nervous System Autoimmune Disease, Experimental/metabolism , Nervous System Autoimmune Disease, Experimental/pathology , Transcriptome
4.
Neuroscience ; 344: 148-156, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28049030

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease in which more than 70% of patients experience visual disturbance as the earliest symptoms. Lysolecithin (LPC)-induced focal demyelination model has been developed to evaluate the effects of different therapies on myelin repair improvement. In this study, the effects of pregabalin administration on myelin repair and glial activation were investigated. Local demyelination was induced by administration of LPC (1%, 2µL) into the rat optic chiasm. Rats underwent daily injection of pregabalin (30mg/kg, i.p) or vehicle. Visual-evoked potentials (VEPs) recordings were performed for evaluating the function of optic pathway on days 3, 7, 14 and 28 post lesions. Myelin specific staining and immunostaining against GFAP and Iba1 were also carried out for assessment of myelination and glial activation respectively. Electrophysiological data indicated that pregabalin administration could significantly reduce the P1-N1 latency and increase the amplitude of VEPs waves compared to saline group. Luxol fast blue staining and immunostaining against PLP, as mature myelin marker, showed that myelin repair was improved in animals received pregabalin treatment. In addition, pregabalin effectively reduced the expression of GFAP and Iba1 as activated glial markers in optic chiasm. The present study indicates that pregabalin administration enhances myelin repair and ameliorates glial activation of optic chiasm following local injection of LPC.


Subject(s)
Myelin Sheath/drug effects , Nervous System Autoimmune Disease, Experimental/drug therapy , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Optic Chiasm/drug effects , Pregabalin/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Demyelinating Autoimmune Diseases, CNS/drug therapy , Demyelinating Autoimmune Diseases, CNS/pathology , Demyelinating Autoimmune Diseases, CNS/physiopathology , Evoked Potentials, Visual/drug effects , Glial Fibrillary Acidic Protein/metabolism , Lysophosphatidylcholines , Male , Microfilament Proteins/metabolism , Myelin Sheath/pathology , Myelin Sheath/physiology , Nervous System Autoimmune Disease, Experimental/pathology , Nervous System Autoimmune Disease, Experimental/physiopathology , Neuroglia/pathology , Neuroglia/physiology , Optic Chiasm/pathology , Optic Chiasm/physiopathology , Random Allocation , Rats, Wistar
5.
J Cardiol ; 68(3): 253-60, 2016 09.
Article in English | MEDLINE | ID: mdl-26475067

ABSTRACT

BACKGROUND: The renin angiotensin system (RAS) plays an important role in the pathogenesis of cardiovascular diseases and inflammation. Myocarditis is an inflammatory disease of the heart, and the role of the RAS in its pathophysiology is unknown. Because the direct renin inhibitor, aliskiren, is thought to block RAS completely, we investigated the cardioprotective effect of aliskiren in mice with experimental autoimmune myocarditis (EAM). METHODS: A cardiac α-myosin heavy chain peptide was injected in mice on days 0 and 7. Aliskiren 25mg/kg per day (n=10) or vehicle (n=10) was administered to EAM mice starting on day 0 and the animals were killed on day 21. RESULTS: Aliskiren significantly prevented the progression of left ventricular wall thickening in EAM hearts compared to the vehicle-treated group. Histologically, the inflammatory cell infiltration and fibrosis area ratios in the aliskiren-treated group were lower than that in the vehicle-treated group. Immunohistochemistry revealed that aliskiren suppressed CD4 positive cell infiltration in EAM hearts compared to vehicle. Moreover, aliskiren decreased mRNA levels of interleukin (IL)-2, interferon-γ, tumor necrosis factor-α, and collagen 1. In vitro study showed that aliskiren inhibited T cell proliferation and IL-2 production induced by myosin stimulation. CONCLUSION: Our results suggest that aliskiren ameliorates EAM by suppressing T-cell activation and inflammatory cytokines, and has potential as a treatment for myocarditis.


Subject(s)
Amides/pharmacology , Antihypertensive Agents/pharmacology , Fumarates/pharmacology , Nervous System Autoimmune Disease, Experimental/drug therapy , Renin/antagonists & inhibitors , Animals , Cytokines/genetics , Cytokines/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , RNA, Messenger/drug effects , RNA, Messenger/metabolism
6.
Ann Neurol ; 72(4): 610-24, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23109155

ABSTRACT

OBJECTIVE: Microvesicles (MVs) have been indicated as important mediators of intercellular communication and are emerging as new biomarkers of tissue damage. Our previous data indicate that reactive microglia/macrophages release MVs in vitro. The aim of the study was to evaluate whether MVs are released by microglia/macrophages in vivo and whether their number varies in brain inflammatory conditions, such as multiple sclerosis (MS). METHODS: Electron and fluorescence microscopy and flow cytometry were used to detect myeloid MVs in the cerebrospinal fluid (CSF) of healthy controls, MS patients, and rodents affected by experimental autoimmune encephalomyelitis (EAE), the animal model of MS. RESULTS: Myeloid MVs were detected in CSF of healthy controls. In relapsing and remitting EAE mice, the concentration of myeloid MVs in the CSF was significantly increased and closely associated with disease course. Analysis of MVs in the CSF of 28 relapsing patients and 28 patients with clinical isolated syndrome from 2 independent cohorts revealed higher levels of myeloid MVs than in 13 age-matched controls, indicating a clinical value of MVs as a companion tool to capture disease activity. Myeloid MVs were found to spread inflammatory signals both in vitro and in vivo at the site of administration; mice impaired in MV shedding were protected from EAE, suggesting a pathogenic role for MVs in the disease. Finally, FTY720, the first approved oral MS drug, significantly reduced the amount of MVs in the CSF of EAE-treated mice. INTERPRETATION: These findings identify myeloid MVs as a marker and therapeutic target of brain inflammation.


Subject(s)
Biomarkers/cerebrospinal fluid , Central Nervous System Diseases/cerebrospinal fluid , Central Nervous System Diseases/drug therapy , Inflammation/cerebrospinal fluid , Inflammation/drug therapy , Spinal Cord/metabolism , Animals , Blotting, Western , Calcium Signaling/physiology , Cell Communication , Cells, Cultured , Encephalitis/cerebrospinal fluid , Encephalitis/pathology , Flow Cytometry , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Multiple Sclerosis/pathology , Nervous System Autoimmune Disease, Experimental/cerebrospinal fluid , Nervous System Autoimmune Disease, Experimental/drug therapy , Neuroglia/metabolism , Neuroglia/physiology , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/physiology
7.
J Nutr Biochem ; 23(11): 1498-507, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22402368

ABSTRACT

Nutraceuticals and phytochemicals are important regulators of human health and diseases. Curcumin is a polyphenolic phytochemical isolated from the rhizome of the plant Curcuma longa (turmeric) that has been traditionally used for the treatment of inflammation and wound healing for centuries. Systematic analyses have shown that curcumin exerts its beneficial effects through antioxidant, antiproliferative and anti-inflammatory properties. We and others have shown earlier that curcumin ameliorates experimental autoimmune encephalomyelitis (EAE) model for multiple sclerosis. In this study, we show that C57BL/6 mice induced to develop EAE express elevated levels of interferon (IFN) γ and interleukin (IL)-17 in the central nervous system (CNS) and lymphoid organs that decreased significantly following in vivo treatment with curcumin. The EAE mice also showed elevated expression of IL-12 and IL-23 that decreased after treatment with curcumin. Ex vivo and in vitro treatment with curcumin resulted in a dose-dependent decrease in the secretion of IFNγ, IL-17, IL-12 and IL-23 in culture. The inhibition of EAE by curcumin was also associated with an up-regulation of IL-10, peroxisome proliferator activated receptor γ and CD4(+)CD25(+-)Foxp3(+) Treg cells in the CNS and lymphoid organs. These findings highlight that curcumin differentially regulates CD4(+) T helper cell responses in EAE.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Curcumin/pharmacology , Encephalomyelitis/drug therapy , Encephalomyelitis/immunology , T-Lymphocytes, Helper-Inducer/drug effects , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis/metabolism , Female , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-17/metabolism , Interleukin-23/metabolism , Mice , Mice, Inbred C57BL , Nervous System Autoimmune Disease, Experimental/drug therapy , Spleen/cytology , Spleen/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
8.
Arthritis Rheum ; 60(10): 3118-27, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19790078

ABSTRACT

OBJECTIVE: Polymyositis and dermatomyositis are chronic inflammatory muscle diseases. Retinoids are compounds that bind to the retinoic acid binding site of retinoic acid receptors and have biologic activities similar to those of vitamin A. Recent studies indicate that retinoids promote Th2 differentiation and suppress Th1 and Th17 differentiation in vitro. The present study was undertaken to examine the effects of a synthetic retinoid, Am80, on experimental autoimmune myositis as well as on Th phenotype development and antibody production. METHODS: Experimental autoimmune myositis was induced in SJL/J mice by immunization with rabbit myosin. Am80 was administered orally once daily. Its effects were evaluated by measurement of the numbers of infiltrating inflammatory cells, production of inflammatory cytokines in muscle, production of Th-specific cytokines by myosin-stimulated splenic T cells, and production of antimyosin antibodies in serum. RESULTS: In mice with experimental autoimmune myositis, orally administered Am80 significantly reduced the number of infiltrating inflammatory cells and the expression of tumor necrosis factor alpha and interleukin-1beta (IL-1beta) in muscle. Moreover, Am80 increased production of interferon-gamma, IL-4, and IL-10, but not IL-17, by myosin-stimulated splenic T cells of mice with experimental autoimmune myositis, suggesting that it could enhance differentiation into Th1 and Th2, but not Th17, in vivo. Am80 also decreased serum levels of IgG2a and IgG2b antimyosin antibodies, but did not affect levels of IgG1 antimyosin antibodies. In addition, it suppressed chemokine expression and activator protein 1 activity in myoblasts in vitro. CONCLUSION: The synthetic retinoid Am80 has an inhibitory effect on experimental autoimmune myositis. It might regulate the development of Th phenotype and antibody production in vivo, in addition to its effects on cytokine and chemokine production.


Subject(s)
Antibodies/metabolism , Cell Differentiation/drug effects , Nervous System Autoimmune Disease, Experimental/immunology , Nervous System Autoimmune Disease, Experimental/prevention & control , Retinoids/pharmacology , T-Lymphocytes, Helper-Inducer/pathology , Animals , Benzoates/pharmacology , Disease Models, Animal , Immunoglobulin G/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Mutant Strains , Myosins/immunology , Nervous System Autoimmune Disease, Experimental/drug therapy , Retinoids/therapeutic use , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Tetrahydronaphthalenes/pharmacology , Th1 Cells/drug effects , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/drug effects , Th2 Cells/metabolism , Th2 Cells/pathology , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Nervenarzt ; 80(2): 155-6, 158-60, 162-5, 2009 Feb.
Article in German | MEDLINE | ID: mdl-19183926

ABSTRACT

Rituximab, a human-mouse chimeric CD20 monoclonal antibody that depletes CD20-positive B cells, has already demonstrated efficacy in hematologic and rheumatologic diseases. Treatment with rituximab results in depletion of CD20-positive cells via multiple mechanisms, including complement-mediated or antibody-dependent cytotoxicity and apoptosis. Recent histopathologic and immunologic studies reveal an influence of B cells on the development and perpetuation of many chronic inflammatory diseases of the nervous system. Promising results with rituximab were already reported in the therapy of myasthenia gravis, immunoneuropathies, neuromyelitis optica, and multiple sclerosis, in which first controlled studies have been recently published. In this review we summarize available data from these reports and also discuss possible underlying molecular mechanisms.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD20/immunology , B-Lymphocytes/immunology , Nervous System Autoimmune Disease, Experimental/drug therapy , Nervous System Autoimmune Disease, Experimental/immunology , Neuroimmunomodulation/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Murine-Derived , B-Lymphocytes/drug effects , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/immunology , Models, Immunological , Neuroimmunomodulation/drug effects , Rituximab
10.
PLoS One ; 4(1): e4151, 2009.
Article in English | MEDLINE | ID: mdl-19127301

ABSTRACT

BACKGROUND: Minocycline is an oral tetracycline derivative with good bioavailability in the central nervous system (CNS). Minocycline, a potent inhibitor of matrix metalloproteinase (MMP)-9, attenuates disease activity in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Potential adverse effects associated with long-term daily minocycline therapy in human patients are concerning. Here, we investigated whether less frequent treatment with long-circulating polyethylene glycol (PEG) minocycline liposomes are effective in treating EAE. FINDINGS: Performing in vitro time kinetic studies of PEG minocycline-liposomes in human peripheral blood mononuclear cells (PBMCs), we determined that PEG minocycline-liposome preparations stabilized with CaCl(2) are effective in diminishing MMP-9 activity. Intravenous injections of PEG minocycline-liposomes every five days were as effective in ameliorating clinical EAE as daily intraperitoneal injections of minocycline. Treatment of animals with PEG minocycline-liposomes significantly reduced the number of CNS-infiltrating leukocytes, and the overall expression of MMP-9 in the CNS. There was also a significant suppression of MMP-9 expression and proteolytic activity in splenocytes of treated animals, but not in CNS-infiltrating leukocytes. Thus, leukocytes gaining access to the brain and spinal cord require the same absolute amount of MMP-9 in all treatment groups, but minocycline decreases the absolute cell number. CONCLUSIONS: Our data indicate that less frequent injections of PEG minocycline-liposomes are an effective alternative pharmacotherapy to daily minocycline injections for the treatment of CNS autoimmune diseases. Also, inhibition of MMP-9 remains a promising treatment target in EAE and patients with MS.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Minocycline/administration & dosage , Polyethylene Glycols/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Liposomes , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Minocycline/therapeutic use , Nervous System Autoimmune Disease, Experimental/drug therapy , Nervous System Autoimmune Disease, Experimental/immunology , Polyethylene Glycols/pharmacology
11.
J Leukoc Biol ; 82(3): 532-41, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17537989

ABSTRACT

Previous studies have shown that cannabinoids have anti-inflammatory and immune-modulating effects, but the precise mechanisms of action remain to be elucidated. In this study, we investigated the effect of JWH 133, a selective agonist for cannabinoid receptor 2, the main receptor expressed on immune cells, in a model of autoimmune disease, experimental autoimmune uveoretinitis (EAU). JWH 133 suppressed EAU in a dose-dependent manner (0.015-15 mg/kg), and the suppressive effect could be achieved in the disease-induction stage and the effector stage. Leukocytes from mice, which had been treated with JWH 133, had diminished responses to retinal peptide and mitogen Con A stimulation in vitro. In vivo JWH 133 treatment also abrogated leukocyte cytokine/chemokine production. Further in vitro studies indicated that JWH 133 down-regulated the TLR4 via Myd88 signal transduction, which may be responsible for its moderate, suppressive effect on antigen presentation. In vivo JWH 133 treatment (1 mg/kg) also suppressed leukocyte trafficking (rolling and infiltration) in inflamed retina as a result of an effect on reducing adhesion molecules CD162 (P-selectin glycoprotein ligand 1) and CD11a (LFA-1) expression on T cells. In conclusion, the cannabinoid agonist JWH 133 has a high in vivo, anti-inflammatory property and may exert its effect via inhibiting the activation and function of autoreactive T cells and preventing leukocyte trafficking into the inflamed tissue.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabinoids/pharmacology , Nervous System Autoimmune Disease, Experimental/immunology , Receptor, Cannabinoid, CB2/agonists , Retinitis/drug therapy , Uveitis/drug therapy , Animals , Antigen-Presenting Cells/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation/drug effects , Chemotaxis, Leukocyte , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , Nervous System Autoimmune Disease, Experimental/drug therapy , Nervous System Autoimmune Disease, Experimental/physiopathology , Receptor, Cannabinoid, CB2/metabolism , Retinitis/immunology , Signal Transduction , T-Lymphocyte Subsets/immunology , Toll-Like Receptor 4/metabolism , Uveitis/immunology
12.
Biochem Pharmacol ; 72(12): 1697-706, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17007821

ABSTRACT

Many reports have shown that cannabinoids might be beneficial in the symptomatic treatment of multiple sclerosis (MS). We have investigated the therapeutic properties of the non-selective cannabinoid receptor agonist WIN-2 as a suppressive drug in the experimental autoimmune encephalomyelitis (EAE) model of MS. In the passive variety of EAE, induced in Lewis rats by adoptive transfer of myelin-reactive T cells, WIN-2 ameliorates the clinical signs and diminishes the cell infiltration of the spinal cord. Due to the involvement of cannabinoids in the regulation of cell death and survival, we investigated the effects of WIN-2 on the encephalitogenic T cell population. WIN-2 induced a profound increase of apoptosis in a dose- and time-dependent manner. The potential involvement of cannabinoid receptors (CB) was investigated by encephalitogenic T cell stimulation in the presence of the CB(1) (SR141716A) and CB(2) (SR144528) antagonists, pertussis toxin (PTX) and the inactive enantiomer WIN-3. WIN-2-induced apoptosis was partially blocked by SR144528 and PTX, whereas, WIN-3 only exerted a mild effect on cell viability. These results point to the partial involvement of CB(2) receptor together with other receptor-independent mechanism or by yet unknown cannabinoid receptors. Moreover, WIN-2 induced the extrinsic pathway of apoptosis, as shown by caspase-10 and -3 activation. These results suggest that cannabinoid-induced apoptosis of encephalitogenic T cells may cooperate in their anti-inflammatory action in EAE models. The partial involvement of CB(2) receptors in WIN-2 action may open new therapeutic doors in the management of MS by non-psychoactive selective cannabinoid agonists.


Subject(s)
Apoptosis/drug effects , Cannabinoids/therapeutic use , Encephalomyelitis/drug therapy , Morpholines/therapeutic use , Multiple Sclerosis/drug therapy , Naphthalenes/therapeutic use , Nervous System Autoimmune Disease, Experimental/drug therapy , Receptor, Cannabinoid, CB2/metabolism , T-Lymphocytes/drug effects , Adoptive Transfer , Animals , Apoptosis/immunology , Benzoxazines , Cannabinoids/administration & dosage , Cannabinoids/pharmacology , Caspases/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Encephalomyelitis/immunology , Encephalomyelitis/metabolism , Injections, Intraperitoneal , Morpholines/administration & dosage , Morpholines/pharmacology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Nervous System Autoimmune Disease, Experimental/immunology , Nervous System Autoimmune Disease, Experimental/metabolism , Rats , Rats, Inbred Lew , Receptor, Cannabinoid, CB2/agonists , T-Lymphocytes/immunology
13.
J Leukoc Biol ; 79(6): 1193-201, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16574770

ABSTRACT

Experimental autoimmune uveoretinitis (EAU) is a T helper type 1 cell-mediated autoimmune disease, which serves as a model of human chronic uveitis. In this model, cells of a monocyte/macrophage lineage and retinal antigen (Ag)-specific T cells infiltrate into the retina and cause inflammatory lesion, where proinflammatory cytokines and various stimuli activate a transcriptional factor, nuclear factor-kappaB (NF-kappaB), which modulates inflammation and enhances immune responses. In the present study, the therapeutic effect of administration of a NF-kappaB inhibitor, pyrrolidine dithiocarbamate (PDTC), was examined in a murine EAU model. It was shown that PDTC ameliorated the clinical symptoms of EAU mice and significantly reduced the histopathological score compared with those in untreated mice. mRNA expressions of tumor necrosis factor alpha and interleukin-1beta were suppressed in eyes of PDTC-treated EAU mice. However, when T cells from PDTC-treated EAU mice, Ag-presenting cells (APC), and the retinal Ag peptides were cocultured, these T cells showed the same level of proliferation as those from control mice. Furthermore, addition of PDTC in the culture of T cells from EAU mice, Ag, and APC completely abrogated the T cell-proliferative response and cytokine production. Pretreatment of Ag-primed T cells or APC with PDTC in vitro also reduced these responses. These results indicate that the inhibitory effect of PDTC is attributed mainly to the suppression of effector-phase responses including inflammation but not to the inhibition of T cell priming. Regulation of NF-kappaB pathway in the lesion could be a novel target for the successful control of uveoretinitis.


Subject(s)
NF-kappa B/antagonists & inhibitors , Nervous System Autoimmune Disease, Experimental/drug therapy , Pyrrolidines/therapeutic use , Retinitis/drug therapy , Thiocarbamates/therapeutic use , Uveitis/drug therapy , Amino Acid Sequence , Animals , Antigen-Presenting Cells/immunology , Autoantigens/chemistry , Autoantigens/immunology , Autoantigens/toxicity , Cell Division/drug effects , Crosses, Genetic , Drug Evaluation, Preclinical , Eye Proteins/chemistry , Eye Proteins/immunology , Female , Gene Expression Regulation/drug effects , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukins/biosynthesis , Interleukins/genetics , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Sequence Data , Peptide Fragments/immunology , Peptide Fragments/toxicity , Protein Transport/drug effects , Pyrrolidines/pharmacology , RNA, Messenger/biosynthesis , Retinol-Binding Proteins/chemistry , Retinol-Binding Proteins/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , Thiocarbamates/pharmacology , Transcription Factor RelA/analysis , Transcription Factor RelA/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
14.
Exp Neurol ; 193(1): 163-71, 2005 May.
Article in English | MEDLINE | ID: mdl-15817275

ABSTRACT

In patients with multiple sclerosis (MS), non-remitting deficits are mainly caused by axonal and neuronal damage. We demonstrated previously that myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis in rats provokes severe axonal and neuronal injury even before clinical manifestation of the disease. In our present study, we investigated effects of simvastatin treatment on degeneration of retinal ganglion cell (RGC) bodies as well as their axons during MOG-induced optic neuritis. Electrophysiological functions of optic nerves and RGCs were analyzed in vivo. Although neuroprotective effects of simvastatin have been demonstrated before in other experimental settings, we did not observe an increase in RGC survival nor an improvement of visual functions. As we could not reproduce the anti-inflammatory effects that were observed under statin therapy in other EAE models, we hypothesize that patients suffering from optic neuritis might not take advantage of simvastatin applications.


Subject(s)
Nervous System Autoimmune Disease, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Optic Neuritis/prevention & control , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/drug effects , Simvastatin/therapeutic use , Animals , Female , Nervous System Autoimmune Disease, Experimental/metabolism , Nervous System Autoimmune Disease, Experimental/pathology , Neuroprotective Agents/pharmacology , Optic Neuritis/metabolism , Optic Neuritis/pathology , Rats , Rats, Inbred BN , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Simvastatin/pharmacology
15.
Mult Scler ; 8(2): 130-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11990870

ABSTRACT

Treatment with paclitaxel by four intraperitoneal injections (20 mg/kg) 1 week apart attenuated clinical signs in a spontaneously demyelinating model, if given with onset of clinical signs. If given at 2 months of age (1 month prior to clinical signs), disease was almost completely prevented The astrogliosis, prominent in our model, was reversed by paditaxel as determined by astrocyte counts and quantitation of GFAP. Electron microscopic examination of affected regions at 2.5 months demonstrated that the myelin was generally normal. By 4 months of age, demyelination was common in the superior cerebellar peduncle, maximal at 6 months, but continued to 8 months. In addition to myelin vacuolation and nude axons, the presence of many thin myelin sheaths suggested remyelination or partial demyelination. Although no evidence of oligodendrocyte loss was seen, nuclear changes were observed. To substantiate that remyelination was occurring, we measured MBP (18.5 kDa), MBP-exon II, Golli-MBP, TP8, Golli-MBP-J37, platelet-derived growth factor alpha (PDGFR alpha) and sonic hedgehog (SHH). Of these TP8, PDGFR alpha and SHH were up-regulated in the untreated transgenic. After paditaxel treatment, MBP-Exon II, TP8, PDGFR alpha and SHH were further up-regulated. We concluded that some of the effects of paditaxel were to stimulate proteins involved in early myelinating events possibly via a signal transduction mechanism.


Subject(s)
Hereditary Central Nervous System Demyelinating Diseases/drug therapy , Multiple Sclerosis , Nervous System Autoimmune Disease, Experimental/drug therapy , Paclitaxel/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/ultrastructure , Biomarkers , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Division/drug effects , Cell Nucleus/ultrastructure , Cerebellum/pathology , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/analysis , Gliosis/drug therapy , Gliosis/genetics , Gliosis/pathology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Injections, Intraperitoneal , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Myelin Proteolipid Protein/genetics , Myelin Sheath/drug effects , Myelin Sheath/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nervous System Autoimmune Disease, Experimental/genetics , Nervous System Autoimmune Disease, Experimental/pathology , Oligodendroglia/pathology , Signal Transduction/drug effects
16.
J Neurol Sci ; 193(1): 7-11, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11718743

ABSTRACT

Experimental autoimmune myositis (EAM) is a good model of human inflammatory myopathy. We induced EAM in SJL/J mice by injection with myosin and treated inflammatory changes with FK506. The mice developed inflammatory changes after the fifth myosin injection. After treatment with FK506, inflammation was suppressed and central nuclei of the muscle fibers increased. These findings indicate that FK506 is effective in the treatment of EAM. The data suggests that FK506 inhibits interaction with calcineurin. Intercellular adhesion molecule-1 (ICAM-1) positive cells were present in the inflammatory and non-inflammatory areas of EAM. The FK506-treated group stained more weakly for ICAM-1 than the untreated EAM group.


Subject(s)
Immunosuppressive Agents/pharmacology , Muscle, Skeletal/drug effects , Myositis/drug therapy , Nervous System Autoimmune Disease, Experimental/drug therapy , Tacrolimus/pharmacology , Animals , Cell Nucleus/drug effects , Cell Nucleus/immunology , Cell Nucleus/pathology , Female , Immunohistochemistry , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred Strains , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myosins/immunology , Myosins/pharmacology , Myositis/immunology , Myositis/metabolism , Nervous System Autoimmune Disease, Experimental/immunology , Nervous System Autoimmune Disease, Experimental/metabolism , Phagocytosis/drug effects , Phagocytosis/immunology , Treatment Outcome
17.
J Neuroimmunol ; 107(1): 83-7, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10808054

ABSTRACT

Recently, it has been shown that inflammatory T-cells in human idiopathic myositis only very rarely undergo spontaneous apoptosis. The animal model of experimental autoimmune myositis (EAM) in the Lewis rat was chosen to investigate whether similar findings hold true in rat muscle and if glucocorticosteroids act by induction of T-cell apoptosis in inflammatory lesions. The rate of spontaneous T-cell apoptosis in rat EAM was low, even in muscle specimens with extensive inflammation. After intravenous glucocorticosteroid pulse therapy we found a dramatic increase in the rate of apoptotic T-cells in the inflamed muscles. Up to 50% of these apoptotic T-cells were CD8 positive apoptotic T-cells. T-cell apoptosis was significantly lower in similarly inflamed muscle specimens of the control group. We suggest that glucocorticosteroids induce apoptosis of endomysial T-cells in human idiopathic polymyositis. Glucocorticosteroid-induced apoptosis may be a candidate mechanism in the termination of inflammatory activity.


Subject(s)
Apoptosis , Glucocorticoids/therapeutic use , Methylprednisolone/therapeutic use , Nervous System Autoimmune Disease, Experimental/drug therapy , Nervous System Autoimmune Disease, Experimental/physiopathology , T-Lymphocytes/physiology , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/physiology , Injections, Intravenous , Muscles/pathology , Muscles/physiopathology , Nervous System Autoimmune Disease, Experimental/pathology , Rats , Rats, Inbred Lew , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL