Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cells ; 12(4)2023 02 08.
Article in English | MEDLINE | ID: mdl-36831209

ABSTRACT

Background: Glatiramer acetate (GA) is a well-established treatment option for patients with clinically isolated syndrome and relapsing-remitting multiple sclerosis (MS) with few side effects. The double transgenic mouse model spontaneous opticospinal encephalomyelitis (OSE), based on recombinant myelin oligodendrocyte glycoprotein35-55 reactive T and B cells, mimicks features of chronic inflammation and degeneration in MS and related disorders. Here, we investigated the effects of prophylactic GA treatment on the clinical course, histological alterations and peripheral immune cells in OSE. Objective: To investigate the effects of prophylactic glatiramer acetate (GA) treatment in a mouse model of spontaneous opticospinal encephalomyelitis (OSE). Methods: OSE mice with a postnatal age of 21 to 28 days without signs of encephalomyelitis were treated once daily either with 150 µg GA or vehicle intraperitoneally (i. p.). The animals were scored daily regarding clinical signs and weight. The animals were sacrificed after 30 days of treatment or after having reached a score of 7.0 due to animal care guidelines. We performed immunohistochemistry of spinal cord sections and flow cytometry analysis of immune cells. Results: Preventive treatment with 150 µg GA i. p. once daily significantly reduced clinical disease progression with a mean score of 3.9 ± 1.0 compared to 6.2 ± 0.7 in control animals (p < 0.01) after 30 d in accordance with positive effects on weight (p < 0.001). The immunohistochemistry showed that general inflammation, demyelination or CD11c+ dendritic cell infiltration did not differ. There was, however, a modest reduction of the Iba1+ area (p < 0.05) and F4/80+ area upon GA treatment (p < 0.05). The immune cell composition of secondary lymphoid organs showed a trend towards an upregulation of regulatory T cells, which lacked significance. Conclusions: Preventive treatment with GA reduces disease progression in OSE in line with modest effects on microglia/macrophages. Due to the lack of established prophylactic treatment options for chronic autoimmune diseases with a high risk of disability, our study could provide valuable indications for translational medicine.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Encephalomyelitis , Mice , Animals , Glatiramer Acetate/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/pathology , Peptides/pharmacology , Inflammation/drug therapy , Mice, Transgenic , Encephalomyelitis/drug therapy , Disease Progression
2.
Proc Natl Acad Sci U S A ; 120(4): e2216941120, 2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36669102

ABSTRACT

In inflammatory neuropathies, oxidative stress results in neuronal and Schwann cell (SC) death promoting early neurodegeneration and clinical disability. Treatment with the short-chain fatty acid propionate showed a significant immunoregulatory and neuroprotective effect in multiple sclerosis patients. Similar effects have been described for patients with chronic inflammatory demyelinating polyneuropathy (CIDP). Therefore, Schwann cell's survival and dorsal root ganglia (DRG) outgrowth were evaluated in vitro after propionate treatment and application of H2O2 or S-nitroso-N-acetyl-D-L-penicillamine (SNAP) to evaluate neuroprotection. In addition, DRG resistance was evaluated by the application of oxidative stress by SNAP ex vivo after in vivo propionate treatment. Propionate treatment secondary to SNAP application on DRG served as a neuroregeneration model. Histone acetylation as well as expression of the free fatty acid receptor (FFAR) 2 and 3, histone deacetylases, neuroregeneration markers, and antioxidative mediators were investigated. ß-hydroxybutyrate was used as a second FFAR3 ligand, and pertussis toxin was used as an FFAR3 antagonist. FFAR3, but not FFAR2, expression was evident on SC and DRG. Propionate-mediated activation of FFAR3 and histone 3 hyperacetylation resulted in increased catalase expression and increased resistance to oxidative stress. In addition, propionate treatment resulted in enhanced neuroregeneration with concomitant growth-associated protein 43 expression. We were able to demonstrate an antioxidative and neuroregenerative effect of propionate on SC and DRG mediated by FFAR3-induced histone acetylases expression. Our results describe a pathway to achieve neuroprotection/neuroregeneration relevant for patients with immune-mediated neuropathies.


Subject(s)
Histones , Propionates , Humans , Propionates/pharmacology , Histones/metabolism , Receptors, G-Protein-Coupled/metabolism , Neuroprotection , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Ganglia, Spinal/metabolism
3.
J Neuroinflammation ; 17(1): 9, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31915017

ABSTRACT

BACKGROUND: The multi-drug resistance transporter ABCG2, a member of the ATP-binding cassette (ABC) transporter family, mediates the efflux of different immunotherapeutics used in multiple sclerosis (MS), e.g., teriflunomide (teri), cladribine, and mitoxantrone, across cell membranes and organelles. Hence, the modulation of ABCG2 activity could have potential therapeutic implications in MS. In this study, we aimed at investigating the functional impact of abcg2 modulation on teri-induced effects in vitro and in vivo. METHODS: T cells from C57BL/6 J wild-type (wt) and abcg2-knockout (KO) mice were treated with teri at different concentrations with/without specific abcg2-inhibitors (Ko143; Fumitremorgin C) and analyzed for intracellular teri concentration (HPLC; LS-MS/MS), T cell apoptosis (annexin V/PI), and proliferation (CSFE). Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6J by active immunization with MOG35-55/CFA. Teri (10 mg/kg body weight) was given orally once daily after individual disease onset. abcg2-mRNA expression (spinal cord, splenic T cells) was analyzed using qRT-PCR. RESULTS: In vitro, intracellular teri concentration in T cells was 2.5-fold higher in abcg2-KO mice than in wt mice. Teri-induced inhibition of T cell proliferation was two fold increased in abcg2-KO cells compared to wt cells. T cell apoptosis demonstrated analogous results with 3.1-fold increased apoptosis after pharmacological abcg2-inhibition in wt cells. abcg2-mRNA was differentially regulated during different phases of EAE within the central nervous system and peripheral organs. In vivo, at a dosage not efficacious in wt animals, teri treatment ameliorated clinical EAE in abcg2-KO mice which was accompanied by higher spinal cord tissue concentrations of teri. CONCLUSION: Functional relevance of abcg2 modulation on teri effects in vitro and in vivo warrants further investigation as a potential determinant of interindividual treatment response in MS, with potential implications for other immunotherapies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/physiology , Crotonates/therapeutic use , Disease Models, Animal , Immunotherapy/methods , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Toluidines/therapeutic use , Animals , Crotonates/pharmacology , Female , Humans , Hydroxybutyrates , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/drug therapy , Nitriles , Rats , T-Lymphocytes/drug effects , Toluidines/pharmacology
4.
Acta Neuropathol ; 138(3): 443-456, 2019 09.
Article in English | MEDLINE | ID: mdl-31030237

ABSTRACT

The limited efficacy of glucocorticoids (GCs) during therapy of acute relapses in multiple sclerosis (MS) leads to long-term disability. We investigated the potential of vitamin D (VD) to enhance GC efficacy and the mechanisms underlying this VD/GC interaction. In vitro, GC receptor (GR) expression levels were quantified by ELISA and induction of T cell apoptosis served as a functional readout to assess synergistic 1,25(OH)2D3 (1,25D)/GC effects. Experimental autoimmune encephalomyelitis (MOG35-55 EAE) was induced in mice with T cell-specific GR or mTORc1 deficiency. 25(OH)D (25D) levels were determined in two independent cohorts of MS patients with stable disease or relapses either responsive or resistant to GC treatment (initial cohort: n = 110; validation cohort: n = 85). Gene expression of human CD8+ T cells was analyzed by microarray (n = 112) and correlated with 25D serum levels. In vitro, 1,25D upregulated GR protein levels, leading to increased GC-induced T cell apoptosis. 1,25D/GC combination therapy ameliorated clinical EAE course more efficiently than respective monotherapies, which was dependent on GR expression in T cells. In MS patients from two independent cohorts, 25D deficiency was associated with GC-resistant relapses. Mechanistic studies revealed that synergistic 1,25D/GC effects on apoptosis induction were mediated by the mTOR but not JNK pathway. In line, 1,25D inhibited mTORc1 activity in murine T cells, and low 25D levels in humans were associated with a reduced expression of mTORc1 inhibiting tuberous sclerosis complex 1 in CD8+ T cells. GR upregulation by 1,25D and 1,25D/GC synergism in vitro and therapeutic efficacy in vivo were abolished in animals with a T cell-specific mTORc1 deficiency. Specific inhibition of mTORc1 by everolimus increased the efficacy of GC in EAE. 1,25D augments GC-mediated effects in vitro and in vivo in a T cell-specific, GR-dependent manner via mTORc1 inhibition. These data may have implications for improvement of anti-inflammatory GC therapy.


Subject(s)
Calcitriol/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Glucocorticoids/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Multiple Sclerosis , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , Signal Transduction/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
5.
J Neuroimmunol ; 328: 78-85, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30623801

ABSTRACT

The murine anti-CD52 antibody, an equivalent of the humanized antibody alemtuzumab, which is successfully used in the treatment of multiple sclerosis, was used to explore a potential neuroprotective effect driven by immune cell derived brain-derived neurotrophic factor (BDNF). Therefore, lineage specific constitutive knock-out mice with a BDNF deficiency in T cells and macrophages were used and compared to treated wildtype mice. Neither therapeutic nor preventive application of the murine anti-CD52 antibody in an animal model of multiple sclerosis, the MOG35-55 EAE, revealed a beneficial contribution of immune cell derived BDNF to the disease outcome. Furthermore, preventive application of the murine anti-CD52 antibody worsened the clinical EAE disease course and could only be overcome by a prolonged recovery phase after treatment and before disease induction.


Subject(s)
Alemtuzumab/pharmacology , Brain-Derived Neurotrophic Factor/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunologic Factors/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/immunology
7.
Brain Res ; 1636: 172-182, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26872595

ABSTRACT

The neuropathology of schizophrenia has been reported to be closely associated with microglial activation. In a previous study, using the prenatal PolyI:C schizophrenia animal model, we showed an increase in cell numbers and a reduction in microglial branching in 30-day-old PolyI:C descendants, which suggests that there is microglial activation during adolescence. To provide more information about the activation state, we aimed to examine the expression levels of Iba1, which was reported to be up-regulated in activated microglia. We used a flow cytometric approach and investigated CD11b and CD45, two additional markers for the identification of microglial cells. We demonstrated that intracellular staining against Iba1 can be used as a reliable flow cytometric method for identification of microglial cells. Prenatal PolyI:C treatment had long-term effects on CD11b and CD45 expression. It also resulted in a trend towards increased Iba1 expression. Imbalance in CD11b, CD45, and Iba1 expression might contribute to impaired synaptic surveillance and enhanced activation/inflammatory activity of microglia in adult offspring.


Subject(s)
Flow Cytometry/methods , Gene Expression Regulation, Developmental/physiology , Interferon Inducers/toxicity , Microglia/pathology , Poly I-C/toxicity , Prenatal Exposure Delayed Effects/pathology , Age Factors , Animals , Animals, Newborn , Body Weight/drug effects , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Leukocyte Common Antigens/metabolism , Male , Mice , Microfilament Proteins/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Sex Characteristics
8.
Immunity ; 43(4): 817-29, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26488817

ABSTRACT

Growing empirical evidence suggests that nutrition and bacterial metabolites might impact the systemic immune response in the context of disease and autoimmunity. We report that long-chain fatty acids (LCFAs) enhanced differentiation and proliferation of T helper 1 (Th1) and/or Th17 cells and impaired their intestinal sequestration via p38-MAPK pathway. Alternatively, dietary short-chain FAs (SCFAs) expanded gut T regulatory (Treg) cells by suppression of the JNK1 and p38 pathway. We used experimental autoimmune encephalomyelitis (EAE) as a model of T cell-mediated autoimmunity to show that LCFAs consistently decreased SCFAs in the gut and exacerbated disease by expanding pathogenic Th1 and/or Th17 cell populations in the small intestine. Treatment with SCFAs ameliorated EAE and reduced axonal damage via long-lasting imprinting on lamina-propria-derived Treg cells. These data demonstrate a direct dietary impact on intestinal-specific, and subsequently central nervous system-specific, Th cell responses in autoimmunity, and thus might have therapeutic implications for autoimmune diseases such as multiple sclerosis.


Subject(s)
Autoimmunity/drug effects , Central Nervous System/immunology , Dietary Fats/pharmacology , Duodenum/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Fatty Acids/pharmacology , Lymphopoiesis/drug effects , T-Lymphocyte Subsets/drug effects , Animals , Dietary Fats/toxicity , Duodenum/metabolism , Duodenum/microbiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acids/chemistry , Fatty Acids/toxicity , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Gene Expression Regulation/immunology , Lauric Acids/toxicity , Liver X Receptors , MAP Kinase Signaling System , Mice , Molecular Weight , Orphan Nuclear Receptors/biosynthesis , Orphan Nuclear Receptors/genetics , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Transcriptome
9.
J Neuroimmunol ; 285: 16-21, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26198914

ABSTRACT

In this study we examined the role of fumaric acid esters (FAE) in a spontaneous and chronic animal model, the opticospinal EAE (OSE). Preventive treatment of dimethylfumarate (DMF) promotes onset of disease in animals treated with high dose DMF. This group also exhibited a significantly exacerbated disease course in a therapeutic treatment as compared to the low dose DMF approach, where less demyelination, macrophage infiltration, and increased Nrf2 expression in the spinal cord were observed. We conclude that low dose DMF treatment is effective in the therapy of the spontaneous opticospinal EAE model and mediates neuroprotective effects via the oxidative stress response pathway.


Subject(s)
Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Fumarates/administration & dosage , Animals , Demyelinating Diseases/drug therapy , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/immunology , Esters , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
Brain ; 133(Pt 8): 2248-63, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20826430

ABSTRACT

Brain-derived neurotrophic factor plays a key role in neuronal and axonal survival. Brain-derived neurotrophic factor is expressed in the immune cells in lesions of experimental autoimmune encephalomyelitis and multiple sclerosis, thus potentially mediating neuroprotective effects. We investigated the functional role of brain-derived neurotrophic factor in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Mice deficient for brain-derived neurotrophic factor in immune cells displayed an attenuated immune response in the acute phase of experimental autoimmune encephalomyelitis, but progressive disability with enhanced axonal loss in the chronic phase of the disease. In mice deficient for central nervous system-derived brain-derived neurotrophic factor via glial fibrillary acidic protein-crescentin-mediated deletion, a more severe course of experimental autoimmune encephalomyelitis and an overall increased axonal loss was observed. In a lentiviral approach, injection of brain-derived neurotrophic factor-overexpressing T cells led to a less severe course of experimental autoimmune encephalomyelitis and direct axonal protection. Our data imply a functional role of brain-derived neurotrophic factor in autoimmune demyelination by mediating axon protection.


Subject(s)
Autoimmunity/physiology , Brain-Derived Neurotrophic Factor/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Acute Disease , Animals , Axons/immunology , Axons/pathology , Brain/immunology , Brain/pathology , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Chronic Disease , Disability Evaluation , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis , Neuroimmunomodulation/physiology , Receptor, trkB/metabolism , Spinal Cord/immunology , Spinal Cord/pathology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...