Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 18(1): 244, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34702288

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is a common concomitant disease of late-onset myasthenia gravis (MG). However, the impacts of DM on the progression of late-onset MG were unclear. METHODS: In this study, we examined the immune response in experimental autoimmune myasthenia gravis (EAMG) rats with DM or not. The phenotype and function of the spleen and lymph nodes were determined by flow cytometry. The serum antibodies, Tfh cells, and germinal center B cells were determined by ELISA and flow cytometry. The roles of advanced glycation end products (AGEs) in regulating Tfh cells were further explored in vitro by co-culture assays. RESULTS: Our results indicated clinical scores of EAMG rats were worse in diabetes rats compared to control, which was due to the increased production of anti-R97-116 antibody and antibody-secreting cells. Furthermore, diabetes induced a significant upregulation of Tfh cells and the subtypes of Tfh1 and Tfh17 cells to provide assistance for antibody production. The total percentages of B cells were increased with an activated statue of improved expression of costimulatory molecules CD80 and CD86. We found CD4+ T-cell differentiation was shifted from Treg cells towards Th1/Th17 in the DM+EAMG group compared to the EAMG group. In addition, in innate immunity, diabetic EAMG rats displayed more CXCR5 expression on NK cells. However, the expression of CXCR5 on NKT cells was down-regulated with the increased percentages of NKT cells in the DM+EAMG group. Ex vivo studies further indicated that Tfh cells were upregulated by AGEs instead of hyperglycemia. The upregulation was mediated by the existence of B cells, the mechanism of which might be attributed the elevated molecule CD40 on B cells. CONCLUSIONS: Diabetes promoted both adaptive and innate immunity and exacerbated clinical symptoms in EAMG rats. Considering the effect of diabetes, therapy in reducing blood glucose levels in MG patients might improve clinical efficacy through suppressing the both innate and adaptive immune responses. Additional studies are needed to confirm the effect of glucose or AGEs reduction to seek treatment for MG.


Subject(s)
Adaptive Immunity/physiology , Diabetes Mellitus, Experimental/immunology , Immunity, Innate/physiology , Inflammation Mediators/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Female , Inflammation Mediators/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Rats , Rats, Inbred Lew , Th17 Cells/immunology , Th17 Cells/metabolism
3.
J Neuroinflammation ; 16(1): 282, 2019 Dec 29.
Article in English | MEDLINE | ID: mdl-31884963

ABSTRACT

BACKGROUND: Recent studies have demonstrated that natural killer (NK) cells can modulate other immune components and are involved in the development or progression of several autoimmune diseases. However, the roles and mechanisms of NK cells in regulating experimental autoimmune myasthenia gravis (EAMG) remained to be illustrated. METHODS: To address the function of NK cells in experimental autoimmune myasthenia gravis in vivo, EAMG rats were adoptively transferred with splenic NK cells. The serum antibodies, and splenic follicular helper T (Tfh) cells and germinal center B cells were determined by ELISA and flow cytometry. The roles of NK cells in regulating Tfh cells were further verified in vitro by co-culturing splenocytes or isolated T cells with NK cells. Moreover, the phenotype, localization, and function differences between different NK cell subtypes were determined by flow cytometry, immunofluorescence, and ex vivo co-culturation. RESULTS: In this study, we found that adoptive transfer of NK cells ameliorated EAMG symptoms by suppressing Tfh cells and germinal center B cells. Ex vivo studies indicated NK cells inhibited CD4+ T cells and Tfh cells by inducing the apoptosis of T cells. More importantly, NK cells could be divided into CXCR5- and CXCR5+ NK subtypes according to the expression of CXCR5 molecular. Compared with CXCR5- NK cells, which were mainly localized outside B cell zone, CXCR5+ NK were concentrated in the B cell zone and exhibited higher expression levels of IL-17 and ICOS, and lower expression level of CD27. Ex vivo studies indicated it was CXCR5- NK cells not CXCR5+ NK cells that suppressed CD4+ T cells and Tfh cells. Further analysis revealed that, compared with CXCR5- NK cells, CXCR5+ NK cells enhanced the ICOS expression of Tfh cells. CONCLUSIONS: These findings highlight the different roles of CXCR5- NK cells and CXCR5+ NK cells. It was CXCR5- NK cells but not CXCR5+ NK cells that suppressed Tfh cells and inhibited the autoimmune response in EAMG models.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Female , Mice , Rats , Rats, Inbred Lew
4.
J Neuroinflammation ; 16(1): 202, 2019 Nov 03.
Article in English | MEDLINE | ID: mdl-31679515

ABSTRACT

BACKGROUND: The thymus plays an essential role in the pathogenesis of myasthenia gravis (MG). In patients with MG, natural regulatory T cells (nTreg), a subpopulation of T cells that maintain tolerance to self-antigens, are severely impaired in the thymuses. In our previous study, upregulated nTreg cells were observed in the thymuses of rats in experimental autoimmune myasthenia gravis after treatment with exosomes derived from statin-modified dendritic cells (statin-Dex). METHODS: We evaluated the effects of exosomes on surface co-stimulation markers and Aire expression of different kinds of thymic stromal cells, including cTEC, mTEC, and tDCs, in EAMG rats. The isolated exosomes were examined by western blot and DLS. Immunofluorescence was used to track the exosomes in the thymus. Flow cytometry and western blot were used to analyze the expression of co-stimulatory molecules and Aire in vivo and in vitro. RESULTS: We confirmed the effects of statin-Dex in inducing Foxp3+ nTreg cells and found that both statin-Dex and DMSO-Dex could upregulate CD40 but only statin-Dex increased Aire expression in thymic stromal cells in vivo. Furthermore, we found that the role of statin-Dex and DMSO-Dex in the induction of Foxp3+ nTreg cells was dependent on epithelial cells in vitro. CONCLUSIONS: We demonstrated that statin-Dex increased expression of Aire in the thymus, which may further promote the Foxp3 expression in the thymus. These findings may provide a new strategy for the treatment of myasthenia gravis.


Subject(s)
Exosomes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myasthenia Gravis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Atorvastatin/pharmacology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/metabolism , Female , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/cytology , Thymus Gland , Transcription Factors/metabolism , AIRE Protein
5.
J Neuroimmunol ; 326: 55-61, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30481614

ABSTRACT

Sulfatides have immunomodulatory functions, and play protective roles in multiple autoimmune diseases. In the present study, we showed that sulfatides ameliorated experimental autoimmune neuritis in Lewis rats induced with bovine peripheral myelin, which was associated with decreased proportions of Th1 and Th17 cells. Furthermore, compared control group, cells from sulfatide-treated rats exhibited lower potential in proliferation and IL-17 secretion in the presence of BPM or ConA in vitro. Moreover, sulfatides also reduced the proportions of NK and NKT cells. In summary, our study indicated that sulfatides might become a new therapeutic agent in Guillain-Barré syndrome in the future.


Subject(s)
Neuritis, Autoimmune, Experimental/immunology , Sulfoglycosphingolipids/pharmacology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Female , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Rats , Rats, Inbred Lew , Th1 Cells/drug effects , Th17 Cells/drug effects
6.
J Neuroinflammation ; 15(1): 51, 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29467007

ABSTRACT

BACKGROUND: With the recognition of the key roles of cellular metabolism in immunity, targeting metabolic pathway becomes a new strategy for autoimmune disease treatment. Guillain-Barré syndrome (GBS) is an acute immune-mediated inflammatory demyelinating disease of the peripheral nervous system, characterized by inflammatory cell infiltration. These inflammatory cells, including activated macrophages, Th1 cells, and Th17 cells, generally undergo metabolic reprogramming and rely mainly on glycolysis to exert functions. This study aimed to explore whether enhanced glycolysis contributed to the pathogenesis of experimental autoimmune neuritis (EAN), a classic model of GBS. METHODS: Preventive and therapeutic treatments with glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), were applied to EAN rats. The effects of treatments were determined by clinical scoring, weighting, and tissue examination. Flow cytometry and ELISA were used to evaluate T cell differentiation, autoantibody level, and macrophage functions in vivo and in vitro. RESULTS: Glycolysis inhibition with 2-DG not only inhibited the initiation, but also prevented the progression of EAN, evidenced by the improved clinical scores, weight loss, inflammatory cell infiltration, and demyelination of sciatic nerves. 2-DG inhibited the differentiation of Th1, Th17, and Tfh cells but enhanced Treg cell development, accompanied with reduced autoantibody secretion. Further experiments in vitro proved glycolysis inhibition decreased the nitric oxide production and phagocytosis of macrophages and suppressed the maturation of dendritic cells (DC). CONCLUSION: The effects of glycolysis inhibition on both innate and adaptive immune responses and the alleviation of animal clinical symptoms indicated that enhanced glycolysis contributed to the pathogenesis of EAN. Glycolysis inhibition may be a new therapy for GBS.


Subject(s)
Glycolysis/physiology , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/metabolism , Animals , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Glycolysis/drug effects , Immunity, Cellular/drug effects , Immunity, Cellular/physiology , Lipopolysaccharides/toxicity , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Neuritis, Autoimmune, Experimental/drug therapy , RAW 264.7 Cells , Rats , Rats, Inbred Lew
7.
Mol Immunol ; 94: 200-208, 2018 02.
Article in English | MEDLINE | ID: mdl-29331804

ABSTRACT

Recent studies have demonstrated the important role of toll-like receptor 9 (TLR9) signalling in autoimmune diseases, but its role in myasthenia gravis (MG) has not been fully established. We show herein that blocking TLR9 signalling via the suppressive oligodeoxynucleotide (ODN) H154 alleviated the symptoms of experimental autoimmune myasthenia gravis (EAMG). With the downregulation of dendritic cells (DCs), TLR9 interruption reduced follicular helper T cells (Tfh) and germinal centre (GC) B cells, leading to decreased antibody production. In addition, TLR9+ B cells as well as total B cells in the spleen were inhibited by H154. These findings highlight the critical role of TLR9 in EAMG and suggest that the inhibition of the TLR9 pathway might be a potential pharmacological strategy for the treatment of myasthenia gravis.


Subject(s)
Immunity, Humoral/drug effects , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Toll-Like Receptor 9/antagonists & inhibitors , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Germinal Center/drug effects , Germinal Center/pathology , Immunity, Humoral/genetics , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 9/genetics
8.
Eur J Pharmacol ; 821: 49-56, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29288118

ABSTRACT

3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase inhibitors (statins) have an immuno-regulatory effect in addition to lowing-lipids. Accumulated evidence showed that the expressions of T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) on natural killer (NK) cells increased in atherosclerotic patients and animal models. In this study, 14 patients treated with rosuvastatin and 12 patients with atorvastatin for more than 3 months were included and 20 patients without statins treatment as control. Both statins treatment reduced the expressions of Tim-3 on NK cells and their subtypes, natural killer T (NKT) cells and CD3+ T cells, and increased the proportions of NKT cells among peripheral blood mononuclear cells, accompanied by the decreased levels of total cholesterol, low density lipoprotein, and increased ratios of high density lipoprotein to cholesterol. These may contribute to the functions of statins in the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Atorvastatin/pharmacology , Hepatitis A Virus Cellular Receptor 2/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Killer Cells, Natural/metabolism , Natural Killer T-Cells/metabolism , Rosuvastatin Calcium/pharmacology , Adult , Aged , CD3 Complex/immunology , Cholesterol/blood , Female , Humans , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
J Neuroimmunol ; 311: 71-78, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28844501

ABSTRACT

Accumulating evidence shows that the immunoproteasome participates in the immune response, beyond its initial role in the protein degradation. Here, we tested the effects of the selective immunoproteasome inhibitor, ONX-0914, on experimental autoimmune myasthenia gravis (EAMG). We found that ONX-0914 ameliorated the severity of ongoing EAMG by reducing the autoantibody affinity, accompanied with decreased Tfh cells and antigen presenting cells. Also it reduced the percentage of Th17 cells and inhibited the secretion of IL-17. Our data indicated ONX-0914 may bring benefit for MG therapy.


Subject(s)
Immunity, Humoral/drug effects , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Oligopeptides/therapeutic use , Proteasome Inhibitors/metabolism , Animals , Antigen-Presenting Cells/pathology , Antigens, CD/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunoglobulin G/blood , Interleukin-17/immunology , Interleukin-17/metabolism , Lymph Nodes/pathology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Inbred Lew , Spleen/drug effects , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/pathology
10.
Neurosci Lett ; 656: 169-176, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28743581

ABSTRACT

We have previously demonstrated that Cysteinyl aspartate-specific proteinase-1 (caspase-1) inhibitor ameliorates experimental autoimmune myasthenia gravis (EAMG) by inhibited cellular immune response, via suppressing DC IL-1 ß, CD4+ T and γdT cells IL-17 pathways. In this study, we investigated the effect of caspase-1 inhibitor on humoral immune response of EAMG and further explore the underlying mechanisms. An animal model of MG was induced by region 97-116 of the rat AChR α subunit (R97-116 peptide) in Lewis rats. Rats were treated with caspase-1 inhibitor Ac-YVAD-cmk intraperitoneally (i.p.) every second day from day 13 after the first immunization. Flow cytometry, western blot, immunofluorescence, and enzyme-linked immunosorbent assay (ELISA) were performed to evaluate the neuroprotective effect of caspase-1 inhibitor on humoral immune response of EAMG. The results showed that caspase-1 inhibitor reduced the relative affinity of anti-R97-116 IgG, suppressed germinal center response, decreased follicular helper T cells, and increased follicular regulatory T cells and regulatory B cells. In addition, we found that caspase-1 inhibitor inhibited humoral immunity response in EAMG rats via suppressing IL-6-STAT3-Bcl-6 pathways. These results suggest that caspase-1 inhibitor ameliorates EAMG by regulating humoral immune response, thus providing new insights into the development of myasthenia gravis and other autoimmune diseases therapies.


Subject(s)
Caspase 1/metabolism , Caspase Inhibitors/therapeutic use , Interleukin-6/metabolism , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Female , Immunity, Humoral , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Rats, Inbred Lew , T-Lymphocytes, Helper-Inducer/immunology
11.
J Neuroimmunol ; 305: 154-161, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284336

ABSTRACT

A growing body of evidence suggests the anti-inflammatory and antitumor effects of parthenolide (PAR). Here we show that PAR treatment inhibits the initiation of experimental autoimmune neuritis (EAN), suppresses the production of TNF-α, IFN-γ, IL-1ß and IL-17, and decreases Th1 and Th17 cells at early time point. However, such anti-inflammatory effect vanishes later and PAR impedes the recovery of EAN in late phase, which is accompanied with inhibited apoptosis of inflammatory cells. Our results indicate that PAR plays dual roles in EAN and it is not proper to be applied in autoimmune diseases of nervous system.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Neuritis, Autoimmune, Experimental/drug therapy , Sesquiterpenes/therapeutic use , Analysis of Variance , Animals , Annexin A5/metabolism , Apoptosis/physiology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Flow Cytometry , Forkhead Box Protein O3/metabolism , Freund's Adjuvant/toxicity , Lymph Nodes/pathology , Mycobacterium tuberculosis , Neuritis, Autoimmune, Experimental/etiology , Rats , Rats, Inbred Lew , Sciatic Nerve/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...