1
|
Zhang H, Luan S, Wang F, Yang L, Chen S, Li Z, Wang X, Wang WP, Chen LQ, Wang Y. The Role of Exosomes in Central Immune Tolerance and Myasthenia Gravis. Immunol Invest 2024:1-23. [PMID: 39680429 DOI: 10.1080/08820139.2024.2440772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
BACKGROUND Immune homeostasis plays a crucial role in immunology andis dependent on both central and peripheral tolerance. Centraltolerance and peripheral tolerance occur in the thymus and thesecondary lymphoid tissues, respectively. Tolerance breakdown andimmune regulation defects can lead to autoimmune disorders. In thisreview article, we aimed to describe the role of exosomes inregulating central tolerance and provide a summary of their effectson the pathogenesis, diagnosis, and therapeutic potential inmyasthenia gravis (MG). METHODS Articles for this review wereidentified using the PubMed database. RESULTS As the primarylymphoid organ, the thymus is responsible for building an immunecompetent, yet self-tolerant of T-cell population. Thymic statesinclude thymoma, thymic hyperplasia, and thymic atrophy, which canexert a significant influence on the central immune tolerance andrepresent specific characteristics of MG. Previous studies have foundthat exosomes derived from human thymic epithelial cells carryantigen-presenting molecules and a wide range of tissue restrictedantigens, which may indicate a vital role of thymic exosomes in MG.Besides, exosomal miRNAs and lncRNAs may also play a critical role inthe pathophysiology of MG. CONCLUSION This review provides thetherapeutic and diagnostic potential of exosomes in MG patients.
Collapse
Affiliation(s)
- Hanlu Zhang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Fuqiang Wang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Lin Yang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Sicheng Chen
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhiyang Li
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xuyang Wang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Wen-Ping Wang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Long-Qi Chen
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yun Wang
- Department of thoracic surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Tang Z, Chen M, Chen C, Fan C, Huang J. BMSCs-Derived Extracellular VesiclemiR-29a-3p Improved the Stability of Rat Myasthenia Gravis by Regulating Treg/Th17 Cells. Immunol Invest 2024; 53:1422-1438. [PMID: 39291784 DOI: 10.1080/08820139.2024.2404629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
INTRODUCTION Myasthenia gravis (MG) is an autoimmune disorder. Microvesicle-derived miRNAs have been implicated in autoimmune diseases. However, the role of microvesicle-derived miR-29a-3p in MG remains poorly understood. This study aimed to investigate the therapeutic effect and mechanism of miR-29a-3p derived from stem cell microvesicles (MVs) on experimental autoimmune myasthenia gravis (EAMG) rats. METHODS EAMG was induced in rats by injection of the subunit of the rat nicotinic anti-acetylcholine receptor (AChR) R97-116 peptide.Besides the control group, EAMG rats were randomly allocated into the EAMG model group, MV group, MV-NC-agomir group, and MV- miR-29a-3p-agomir group. RESULTS Our results found that BMSCs-MV promoted miR-29a-3p expression in gastrocnemius of EAMG rats. Bone marrow mesenchymal stem cells (BMSCs) derived microvesicle miR-29a-3p improved the hanging ability and swimming time of EMGA rats and weakened the degree of muscle fiber atrophy. Furthermore, microvesicles from miR-29a-3p overexpressing BMSCs reduced the content of AchR-Ab in the serum of EAMG rats. BMSC-derived microvesicle miR-29a-3p further suppressed the expression of IFN-γ and enhanced the IL-4 and IL-10 in the serum of EAMG rats by restoring the Th17/Treg cells balance. DISCUSSION BMSCs-derived microvesicle miR-29a-3p improved the stability of rat myasthenia gravis by regulating Treg/Th17 cells. It may be an effective treatment for MG.
Collapse
Affiliation(s)
- Zhongben Tang
- Department of Thoracic, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Meiqiu Chen
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Chen Chen
- Department of Thoracic, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Chao Fan
- Department of Thoracic, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Jiaxian Huang
- Graduate School, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
3
|
Theissen L, Schroeter CB, Huntemann N, Räuber S, Dobelmann V, Cengiz D, Herrmann A, Koch-Hölsken K, Gerdes N, Hu H, Mourikis P, Polzin A, Kelm M, Hartung HP, Meuth SG, Nelke C, Ruck T. Recombinant Acetylcholine Receptor Immunization Induces a Robust Model of Experimental Autoimmune Myasthenia Gravis in Mice. Cells 2024; 13:508. [PMID: 38534352 DOI: 10.3390/cells13060508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/25/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Myasthenia gravis (MG) is a prototypical autoimmune disease of the neuromuscular junction (NMJ). The study of the underlying pathophysiology has provided novel insights into the interplay of autoantibodies and complement-mediated tissue damage. Experimental autoimmune myasthenia gravis (EAMG) emerged as a valuable animal model, designed to gain further insight and to test novel therapeutic approaches for MG. However, the availability of native acetylcholine receptor (AChR) protein is limited favouring the use of recombinant proteins. To provide a simplified platform for the study of MG, we established a model of EAMG using a recombinant protein containing the immunogenic sequence of AChR in mice. This model recapitulates key features of EAMG, including fatigable muscle weakness, the presence of anti-AChR-antibodies, and engagement of the NMJ by complement and a reduced NMJ density. Further characterization of this model demonstrated a prominent B cell immunopathology supported by T follicular helper cells. Taken together, the herein-presented EAMG model may be a valuable tool for the study of MG pathophysiology and the pre-clinical testing of therapeutic applications.
Collapse
Affiliation(s)
- Lukas Theissen
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Niklas Huntemann
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Saskia Räuber
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Vera Dobelmann
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Derya Cengiz
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Alexander Herrmann
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Kathrin Koch-Hölsken
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Norbert Gerdes
- Department of Cardiology, Pulmonolgy and Vascular Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Hao Hu
- Department of Cardiology, Pulmonolgy and Vascular Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonolgy and Vascular Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonolgy and Vascular Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonolgy and Vascular Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
- Brain and Mind Center, University of Sidney, Sidney NSW 2050, Australia
- Department of Neurology, Palacky University Olomouc, 77146 Olomouc, Czech Republic
| | - Sven G Meuth
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany
| |
Collapse
|
4
|
Sun J, Xie Q, Sun M, Zhang W, Wang H, Liu N, Wang M. Curcumin protects mice with myasthenia gravis by regulating the gut microbiota, short-chain fatty acids, and the Th17/Treg balance. Heliyon 2024; 10:e26030. [PMID: 38420408 PMCID: PMC10900935 DOI: 10.1016/j.heliyon.2024.e26030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Curcumin is widely used as a traditional drug in Asia. Interestingly, curcumin and its metabolites have been demonstrated to influence the microbiota. However, the effect of curcumin on the gut microbiota in patients with myasthenia gravis (MG) remains unclear. This study aimed to investigate the effects of curcumin on the gut microbiota community, short-chain fatty acids (SCFAs) levels, intestinal permeability, and Th17/Treg balance in a Torpedo acetylcholine receptor (T-AChR)-induced MG mouse model. The results showed that curcumin significantly alleviated the clinical symptoms of MG mice induced by T-AChR. Curcumin modified the gut microbiota composition, increased microbial diversity, and, in particular, reduced endotoxin-producing Proteobacteria and Desulfovibrio levels in T-AChR-induced gut dysbiosis. Moreover, we found that curcumin significantly increased fecal butyrate levels in mice with T-AChR-induced gut dysbiosis. Butyrate levels increased in conjunction with the increase in butyrate-producing species such as Oscillospira, Akkermansia, and Allobaculum in the curcumin-treated group. In addition, curcumin repressed the increased levels of lipopolysaccharide (LPS), zonulin, and FD4 in plasma. It enhanced Occludin expression in the colons of MG mice induced with T-AChR, indicating dramatically alleviated gut permeability. Furthermore, curcumin treatment corrected T-AChR-induced imbalances in Th17/Treg cells. In summary, curcumin may protect mice against myasthenia gravis by modulating both the gut microbiota and SCFAs, improving gut permeability, and regulating the Th17/Treg balance. This study provides novel insights into curcumin's clinical value in MG therapy.
Collapse
Affiliation(s)
- Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Qinfang Xie
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Wenjing Zhang
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, Qinghai, 810007, China
| | - Hongxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, China
| |
Collapse
|
5
|
Sun J, Sun M, Li X, Xie Q, Zhang W, Wang M. MicroRNA‑155‑5p affects regulatory T cell activation and immunosuppressive function by targeting BCL10 in myasthenia gravis. Exp Ther Med 2024; 27:6. [PMID: 38223327 PMCID: PMC10785013 DOI: 10.3892/etm.2023.12293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 10/20/2023] [Indexed: 01/16/2024] Open
Abstract
The imbalance in immune homeostasis plays a crucial role in the pathogenesis of myasthenia gravis (MG). MicroRNAs (miRs) have been identified as key regulators of immune homeostasis. B-cell lymphoma/leukemia 10 (BCL10) has been implicated in the activation and suppressive function of regulatory T cells (Tregs). This study aimed to investigate the potential role of miR-155-5p in modulating the activation and function of Tregs in MG. To achieve this objective, blood samples were collected from MG patients to assess the expression levels of miR-155-5p and BCL10, as well as the proportion of circulating Tregs, in comparison to healthy controls. The correlation between miR-155-5p and BCL10 levels was evaluated in human samples. The expression levels of miR-155-5p and the numbers of circulating Tregs were also examined in an animal model of experimental autoimmune MG (EAMG). A dual-luciferase reporter assay was used to verify whether miR-155-5p can target BCL10. To determine the regulatory function of BCL10 in Tregs, CD4+ CD25+ Tregs were transfected with either small interfering-BCL10 or miR-155-5p inhibitor, and the expression levels of the anti-inflammatory cytokine IL-10 and transcription factors Foxp3, TGF-β1, CTLA4, and ICOS were measured. The results demonstrated that the expression level of miR-155-5p was significantly higher in patients with MG compared with that in healthy controls, whereas the expression level of BCL10 was significantly decreased in patients with MG. Furthermore, there was a significant negative correlation between the expression levels of miR-155-5p and BCL10. The number of circulating Tregs was significantly reduced in patients with MG and in the spleen of rats with EAMG compared with that in the corresponding control groups. The dual-luciferase reporter assay demonstrated that miR-155-5p could target BCL10. The Tregs transfected with si-BCL10 demonstrated significant decreases in the protein levels of TGF-β1 and IL-10, as well as in the mRNA expression levels of Foxp3, TGF-β1, CTLA-4 and ICOS. Conversely, the Tregs transfected with the miR-155-5p inhibitor exhibited a substantial increase in these protein and mRNA expression levels compared with their respective control groups. Furthermore, the knockdown of BCL10 exhibited a decline in the suppressive efficacy of Tregs on the proliferation of CD4+ T cells. Conversely, the suppression of miR-155-5p expression attenuated the inhibition of the BCL10 gene, potentially causing an indirect influence on the suppressive capability of Tregs on the proliferation of CD4+ T cells. BCL10 was thus found to contribute to the activation and immunosuppressive function of Tregs. In summary, the present study demonstrated that miR-155-5p inhibited the activation and immunosuppressive function of Tregs by targeting BCL10, which may be used as a future potential target for the treatment of MG.
Collapse
Affiliation(s)
- Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xiaoling Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Qinfang Xie
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Wenjing Zhang
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
6
|
Sun J, Chen J, Xie Q, Sun M, Zhang W, Wang H, Liu N, Wang Q, Wang M. Sodium butyrate alleviates R97-116 peptide-induced myasthenia gravis in mice by improving the gut microbiota and modulating immune response. J Inflamm (Lond) 2023; 20:37. [PMID: 37924056 PMCID: PMC10625296 DOI: 10.1186/s12950-023-00363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
Fermented butyrate exhibits an anti-inflammatory response to maintain immune homeostasis within the gut. However, the effect and underlying mechanism of butyrate on myasthenia gravis (MG) remain unclear. The changes in the gut microbiota and fecal contents of SCFAs in MG patients were examined. R97-116 peptide was used to induce the experimental autoimmune myasthenia gravis (EAMG) mice and sodium butyrate (NaB) was gavaged to the EAMG mice. Gut microbiota, the frequency of Th1, Th17, Treg, Tfh, and B cells, the levels of IFN-γ, IL-17 A, IL-10, IL-21, and anti-R97-116 IgG, RNA-seq of total B cells in the spleen were explored by metagenomics, flow cytometry, ELISA, and transcriptomics. A significant reduction in SCFA-producing bacteria including Butyricimonas synergistica and functional modules including butyrate synthesis/production II was observed in MG patients and fecal SCFAs detection confirmed the increase. The EAMG mice were successfully constructed and NaB supplementation has changed the composition and function of the gut microbiota. The numbers of Th1, Th17, Tfh, and B cells were significantly increased while that of Treg cells was obviously decreased in EAMG mice compared with controls. Interestingly, NaB treatment has reduced the amounts of Th17, Tfh, and B cells but increased that of Treg cells. Accordingly, the levels of IL-17 A, IL-21, and IgG were increased while IL-10 was decreased in EAMG mice. However, NaB treatment reduced IL-17 A and IL-21 but increased that of IL-10. RNA-seq of B cells has revealed 4577 deferentially expressed genes (DEGs), in which 1218 DEGs were up-regulated while 3359 DEGs were down-regulated in NaB-treated EAMG mice. GO enrichment and KEGG pathway analysis unveiled that the function of these DEGs was mainly focused on immunoglobulin production, mitochondrial respiratory chain complex, ribosome, oxidative phosphorylation, and CNS diseases including amyotrophic lateral sclerosis. We have found that butyrate was significantly reduced in MG patients and NaB gavage could evidently improve MG symptoms in EAMG mice by changing the gut microbiota, regulating the immune response, and altering the gene expression and function of B cells, suggesting NaB might be a potential immunomodulatory supplement for MG drugs.
Collapse
Affiliation(s)
- Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Juanjuan Chen
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Qinfang Xie
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wenjing Zhang
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, 810007, China
| | - Hongxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
7
|
Weng S, Huang L, Cai B, He L, Wen S, Li J, Zhong Z, Zhang H, Huang C, Yang Y, Jiang Q, Liu F. Astragaloside IV ameliorates experimental autoimmune myasthenia gravis by regulating CD4 + T cells and altering gut microbiota. Chin Med 2023; 18:97. [PMID: 37542273 PMCID: PMC10403896 DOI: 10.1186/s13020-023-00798-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Myasthenia gravis (MG) is an antibody-mediated autoimmune disease and its pathogenesis is closely related to CD4 + T cells. In recent years, gut microbiota is considered to play an important role in the pathogenesis of MG. Astragaloside IV (AS-IV) is one of the main active components extracted from Astragalus membranaceus and has immunomodulatory effects. To study the immunomodulatory effect of AS-IV and the changes of gut microbiota on experimental autoimmune myasthenia gravis (EAMG) mice, we explore the possible mechanism of AS-IV in improving MG. METHODS In this study, network pharmacology was utilized to screen the crucial targets of AS-IV in the treatment of MG. Subsequently, a Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed to identify potential pathways through which AS-IV acts against MG. Furthermore, experimental investigations were conducted to validate the underlying mechanism of AS-IV in MG treatment. Before modeling, 5 mice were randomly selected as the control group (CFA group), and the other 10 were induced to EAMG model. These mice were randomly divided into EAMG group and EAMG + AS-IV group, n = 5/group. In EAMG + AS-IV group, AS-IV was administered by gavage. CFA and EAMG groups were given the same volume of PBS. Body weight, grip strength and clinical symptoms were assessed and recorded weekly. At the last administration, the feces were collected for 16S RNA microbiota analysis. The levels of Treg, Th1 and Th17 cells in spleen and Th1 and Th17 cells in thymus were detected by flow cytometry. The levels of IFN-γ, IL-17 and TGF-β in serum were measured by ELISA. Furthermore, fecal microbial transplantation (FMT) experiments were performed for exploring the influence of changed intestinal flora on EAMG. After EAMG model was induced, the mice were treated with antibiotics daily for 4 weeks to germ-free. Then germ-free EAMG mice were randomly divided into two groups: FMT EAMG group, FMT AS-IV group, n = 3/group. Fecal extractions from EAMG and EAMG + AS-IV groups as gathered above were used to administered daily to the respective groups for 4 weeks. Body weight, grip strength and clinical symptoms were assessed and recorded weekly. The levels of Treg, Th1 and Th17 cells in spleen and Th1 and Th17 cells in thymus were detected at the last administration. The levels of IFN-γ, IL-17 and TGF-β in serum were measured by ELISA. RESULTS The network pharmacology and KEGG pathway analysis revealed that AS-IV regulates T cell pathways, including T cell receptor signaling pathway and Th17 cell differentiation, suggesting its potential in improving MG. Further experimental verification demonstrated that AS-IV administration improved muscle strength and body weight, reduced the level of Th1 and Th17 cells, enhanced the level of Treg cells, and resulted in alterations of the gut microbiota, including changes in beta diversity, the Firmicutes/Bacteroidetes (F/B) ratio, and the abundance of Clostridia in EAMG mice. We further conducted FMT tests and demonstrated that the EAMG Abx-treated mice which were transplanted the feces of mice treated with AS-IV significantly alleviated myasthenia symptoms, reduced Th1 and Th17 cells levels, and increased Treg cell levels. CONCLUSION This study speculated that AS-IV ameliorates EAMG by regulating CD4 + T cells and altering the structure and species of gut microbiota of EAMG.
Collapse
Affiliation(s)
- Senhui Weng
- Department of Spleen and Stomach Diseases, Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Yuexiu District, Guangzhou, 510120, China
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Linwen Huang
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Bingxing Cai
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Long He
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
- Department of Spleen and Stomach Diseases, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Shuting Wen
- Department of Spleen and Stomach Diseases, Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Yuexiu District, Guangzhou, 510120, China
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Jinghao Li
- Department of Traditional Chinese Medicine of the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528000, China
| | - Zhuotai Zhong
- Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Haiyan Zhang
- Department of Spleen and Stomach Diseases, Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Yuexiu District, Guangzhou, 510120, China
| | - Chongyang Huang
- Department of Spleen and Stomach Diseases, Guangdong Provincial Hospital of Chinese Medicine, No. 111 Dade Road, Yuexiu District, Guangzhou, 510120, China
| | - Yunying Yang
- Department of Spleen and Stomach Diseases, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Airport Road, Baiyun District, Guangzhou, 510422, China
| | - Qilong Jiang
- Department of Spleen and Stomach Diseases, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Airport Road, Baiyun District, Guangzhou, 510422, China.
| | - Fengbin Liu
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, No.12 Airport Road, Baiyun District, Guangzhou, 510422, China.
- Department of Spleen and Stomach Diseases, First Affiliated Hospital of Guangzhou University of Chinese Medicine, No.16 Airport Road, Baiyun District, Guangzhou, 510422, China.
- Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 2, Helongqi Road, Renhe Town, Baiyun District, Guangzhou, 510000, China.
| |
Collapse
|
8
|
Cabral-Marques O, Moll G, Catar R, Preuß B, Bankamp L, Pecher AC, Henes J, Klein R, Kamalanathan AS, Akbarzadeh R, van Oostveen W, Hohberger B, Endres M, Koolmoes B, Levarht N, Postma R, van Duinen V, van Zonneveld AJ, de Vries-Bouwstra J, Fehres C, Tran F, do Vale FYN, da Silva Souza KB, Filgueiras IS, Schimke LF, Baiocchi GC, de Miranda GC, da Fonseca DLM, Freire PP, Hackel AM, Grasshoff H, Stähle A, Müller A, Dechend R, Yu X, Petersen F, Sotzny F, Sakmar TP, Ochs HD, Schulze-Forster K, Heidecke H, Scheibenbogen C, Shoenfeld Y, Riemekasten G. Autoantibodies targeting G protein-coupled receptors: An evolving history in autoimmunity. Report of the 4th international symposium. Autoimmun Rev 2023; 22:103310. [PMID: 36906052 DOI: 10.1016/j.autrev.2023.103310] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
G protein-coupled receptors (GPCR) are involved in various physiological and pathophysiological processes. Functional autoantibodies targeting GPCRs have been associated with multiple disease manifestations in this context. Here we summarize and discuss the relevant findings and concepts presented in the biennial International Meeting on autoantibodies targeting GPCRs (the 4th Symposium), held in Lübeck, Germany, 15-16 September 2022. The symposium focused on the current knowledge of these autoantibodies' role in various diseases, such as cardiovascular, renal, infectious (COVID-19), and autoimmune diseases (e.g., systemic sclerosis and systemic lupus erythematosus). Beyond their association with disease phenotypes, intense research related to the mechanistic action of these autoantibodies on immune regulation and pathogenesis has been developed, underscoring the role of autoantibodies targeting GPCRs on disease outcomes and etiopathogenesis. The observation repeatedly highlighted that autoantibodies targeting GPCRs could also be present in healthy individuals, suggesting that anti-GPCR autoantibodies play a physiologic role in modeling the course of diseases. Since numerous therapies targeting GPCRs have been developed, including small molecules and monoclonal antibodies designed for treating cancer, infections, metabolic disorders, or inflammatory conditions, anti-GPCR autoantibodies themselves can serve as therapeutic targets to reduce patients' morbidity and mortality, representing a new area for the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Otávio Cabral-Marques
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil; Laboratory of Medical Investigation 29, University of São Paulo School of Medicine, São Paulo, Brazil; Department of Pharmacy and Postgraduate Program of Health and Science, Federal University of Rio Grande do Norte, Natal, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil.
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany; BIH Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), all Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | - Beate Preuß
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Lukas Bankamp
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Ann-Christin Pecher
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Joerg Henes
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - A S Kamalanathan
- Centre for BioSeparation Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Reza Akbarzadeh
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Wieke van Oostveen
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt- Universität zu Berlin, Department of Neurology with Experimental Neurology, Berlin, Germany.; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Berlin, Germany; Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Stroke Research Berlin, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), Partner Site Berlin, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Bryan Koolmoes
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Nivine Levarht
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Rudmer Postma
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Vincent van Duinen
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- LUMC, Department of Internal Medicine (Nephrology), Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden, the Netherlands
| | - Jeska de Vries-Bouwstra
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Cynthia Fehres
- Leiden University Medical Center (LUMC), Department of Rheumatology, Leiden, the Netherlands
| | - Florian Tran
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Fernando Yuri Nery do Vale
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Igor Salerno Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gabriela Crispim Baiocchi
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gustavo Cabral de Miranda
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Dennyson Leandro Mathias da Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Paula Paccielli Freire
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexander M Hackel
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Hanna Grasshoff
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Anja Stähle
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Antje Müller
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, A collaboration of Max Delbruck Center for Molecular Medicine and Charité Universitätsmedizin, and HELIOS Clinic, Department of Cardiology and Nephrology, Berlin 13125, Germany
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel (RCB), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel (RCB), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Franziska Sotzny
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Berlin, Germany
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, NY, USA
| | - Hans D Ochs
- University of Washington School of Medicine and Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Berlin, Germany
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
9
|
Saifina LF, Abdalla M, Gubaidullina LM, Zueva IV, Eltayb WA, El-Arabey AA, Kharlamova AD, Lenina OA, Semenov VE, Petrov KA. Novel slow-binding reversible acetylcholinesterase inhibitors based on uracil moieties for possible treatment of myasthenia gravis and protection from organophosphate poisoning. Eur J Med Chem 2023; 246:114949. [PMID: 36462442 DOI: 10.1016/j.ejmech.2022.114949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
A series of new compounds in which uracil and 3,6-dimethyluracil moieties are bridged with different spacers were prepared and evaluated in vitro for the acetyl- and butyrylcholinesterase (AChE and BChE) inhibitory activities. These bisuracils are shown to be very effective inhibitors of AChE, inhibiting the enzyme at nano- and lower molar concentrations with extremely high selectivity for AChE vs. BChE. Kinetic analysis showed that the lead compound 2h acts as a slow-binding inhibitor of AChE and possess a long drug-target residence time (τ = 1/koff = 18.6 ± 7.5 min). Moreover, compound 2h ameliorated muscle weakness in myasthenia gravis rat model with a lower effective dose and longer lasting effect than pyridostigmine bromide. Besides, it was shown that compound 2h has an effect of increasing efficiency of antidotal therapy as a pretreatment for poisoning by organophosphates.
Collapse
Affiliation(s)
- Liliya F Saifina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia
| | - Mohnad Abdalla
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, China
| | - Liliya M Gubaidullina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia
| | - Irina V Zueva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia
| | - Wafa Ali Eltayb
- Biotechnology Department, Faculty of Science and Technology, Shendi University, Shendi, 11111, Sudan
| | - Amr Ahmed El-Arabey
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, 11751, Egypt
| | - Alexandra D Kharlamova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia
| | - Oksana A Lenina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia
| | - Vyacheslav E Semenov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia.
| | - Konstantin A Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov str., 8, Kazan, 420088, Russia; Kazan Federal University, 18 Kremlyovskaya str, Kazan, 420008, Russia
| |
Collapse
|
10
|
Exploring the Potential Mechanism of Qi-Shen-Di-Huang Drug Formulary for Myasthenia Gravis (MG) based on UHPLC-QE-MS Network Pharmacology and Molecular Docking Techniques. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7416448. [PMID: 36225188 PMCID: PMC9550457 DOI: 10.1155/2022/7416448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/18/2022] [Indexed: 11/14/2022]
Abstract
Myasthenia gravis (MG) is a rare and refractory autoimmune disease, and Qi Shen Di Huang (QSDH) drug formulary is an in-hospital herbal decoction with proven clinical efficacy in treating MG. Currently, most of the research on the QSDH drug formulary has concentrated on its clinical efficacy, and there is a lack of systematic study on the material basis. The active compounds and their mechanism of action have not been entirely determined. Therefore, this study sought to identify the active compounds in the QSDH drug formulary and analyze the key targets and potential mechanisms. We used ultra-performance liquid chromatography Q Exactive-mass spectrometry (UHPLC-QE-MS) and Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) database to identify and screen 85 active ingredients corresponding to 59 potential targets (17 herbs) associated with myasthenia gravis, and further identified AKT1 as the primary core target and the PI3K/AKT signaling pathway as the most substantial enriched pathway. Molecular docking and UPLC-MS analysis identified quercetin, luteolin, wogonin, kaempferol, laccasein, and epigallocatechin gallate are the core compounds of the QSDH drug formulary. In vivo rat studies showed that the QSDH drug formulary reduced Lennon's clinical score and decreased acetylcholine receptor antibody levels in peripheral blood rats with experimental autoimmune myasthenia gravis. In addition, the QSDH drug formulary downregulated P-PI3K/PI3K and P-Akt/Akt protein expression. Collectively, these findings describe the role and potential mechanism of the QSDH drug formulary in the treatment of MG, which exerts potential value by acting on AKT targets and regulating the PI3K/AKT signaling pathway and providing a theoretical reference for QSDH drug formulary application in the clinical treatment of MG.
Collapse
|
11
|
Song J, Zhao R, Yan C, Luo S, Xi J, Ding P, Li L, Hu W, Zhao C. A Targeted Complement Inhibitor CRIg/FH Protects Against Experimental Autoimmune Myasthenia Gravis in Rats via Immune Modulation. Front Immunol 2022; 13:746068. [PMID: 35154091 PMCID: PMC8825366 DOI: 10.3389/fimmu.2022.746068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Antibody-induced complement activation may cause injury of the neuromuscular junction (NMJ) and is thus considered as a primary pathogenic factor in human myasthenia gravis (MG) and animal models of experimental autoimmune myasthenia gravis (EAMG). In this study, we tested whether CRIg/FH, a targeted complement inhibitor, could attenuate NMJ injury in rat MG models. We first demonstrated that CRIg/FH could inhibit complement-dependent cytotoxicity on human rhabdomyosarcoma TE671 cells induced by MG patient-derived IgG in vitro. Furthermore, we investigated the therapeutic effect of CRIg/FH in a passive and an active EAMG rodent model. In both models, administration of CRIg/FH could significantly reduce the complement-mediated end-plate damage and suppress the development of EAMG. In the active EAMG model, we also found that CRIg/FH treatment remarkably reduced the serum concentration of autoantibodies and of the cytokines including IFN-γ, IL-2, IL-6, and IL-17, and upregulated the percentage of Treg cells in the spleen, which was further verified in vitro. Therefore, our findings indicate that CRIg/FH may hold the potential for the treatment of MG via immune modulation.
Collapse
Affiliation(s)
- Jie Song
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Rui Zhao
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Jianying Xi
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| |
Collapse
|
12
|
Song J, Yang J, Jing S, Yan C, Huan X, Chen S, Zhong H, Lu J, Xi J, Luo L, Chen X, Wang Z, Zhao C, Chu M, Luo S. Berberine attenuates experimental autoimmune myasthenia gravis via rebalancing the T cell subsets. J Neuroimmunol 2022; 362:577787. [PMID: 34923373 DOI: 10.1016/j.jneuroim.2021.577787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 01/18/2023]
Abstract
Myasthenia Gravis (MG) is a T cell-driven, autoantibody-mediated disease. Here we show that oral Berberine (BBR) ameliorated clinical symptoms of experimental autoimmune myasthenia gravis(EAMG) rat model via decreasing the frequencies of Th1, Th17, Th1/17 cell subsets. JAK-STAT pathway was highlighted by transcriptomic analysis with EAMG mononuclear cells (MNCs). Surface plasmon resonance identified ligand binding interaction between BBR and JAK2, and electrostatic interaction was proposed by molecular dynamic simulation. Reduced phosphorylated JAK1/2/3 and STAT1/3 in MNCs from BBR-fed EAMG rats were demonstrated. These results suggest that BBR might improve EAMG by rebalancing T cell subsets through targeting JAK-STAT pathway.
Collapse
Affiliation(s)
- Jie Song
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jie Yang
- Department of Neurology, Wuhan No.1 Hospital, Wuhan, Hubei 430020, China
| | - Sisi Jing
- Department of Neurology, Jing'an District Center Hospital of Shanghai, Shanghai 200040, China
| | - Chong Yan
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Xiao Huan
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Sheng Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Huahua Zhong
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jun Lu
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jianying Xi
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Lijun Luo
- Department of Neurology, Wuhan No.1 Hospital, Wuhan, Hubei 430020, China
| | - Xi Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University. NHC Key Laboratory of Medical Immunology (Peking University), Beijing 100191, China
| | - Ziyuan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University. NHC Key Laboratory of Medical Immunology (Peking University), Beijing 100191, China
| | - Chongbo Zhao
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China
| | - Ming Chu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou 350001, China; Department of Immunology, School of Basic Medical Sciences, Peking University. NHC Key Laboratory of Medical Immunology (Peking University), Beijing 100191, China.
| | - Sushan Luo
- Department of Neurology, Huashan hospital Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai 200040, China.
| |
Collapse
|
13
|
Wang Q, Liu Y, Kuang S, Li R, Weng N, Zhou Z. miR-181a Ameliorates the Progression of Myasthenia Gravis by Regulating TRIM9. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1303375. [PMID: 34925522 PMCID: PMC8677396 DOI: 10.1155/2021/1303375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022]
Abstract
Abnormally activated CD4+ T cells are considered to be an important factor in the pathogenesis of myasthenia gravis (MG). In the pathogenesis of MG, the imbalance of proinflammatory cytokines and immune cells maintains the imbalance of immune response and inflammatory microenvironment. Studies have shown that miRNA is involved in the pathogenesis of MG. In our experiment, we extracted peripheral blood mononuclear cells (PBMCs) from MG patients and detected the expression of miR-181a and TRIM9 in PBMCs by qRT-PCR. In vitro experiments were conducted to explore the regulatory mechanism of miR-181a on target genes and its influence on inflammatory factors related to MG disease. Experimental autoimmune myasthenia gravis (EAMG) model mice are established, and the effects of miR-181a on EAMG symptoms and inflammatory factors are explored through in vivo experiments. According to a total of 40 EAMG mice that were successfully modeled, all EAMG mice showed symptoms of muscle weakness; their diet was reduced; their weight gain was slow; and even weight loss occurred. In MG patients and EAMG mice, the expression of miR-181a was low and TRIM9 was highly expressed. Bioinformatics website and dual-luciferase report analysis of miR-181a had a targeting relationship with TRIM9, and miR-181a could target the expression of TRIM9. After upregulating miR-181a or interfering with TRIM9, serum miR-181a in EAMG mice was significantly upregulated; TRIM9 was significantly downregulated; its clinical symptoms were reduced; and the expression of inflammatory factors was reduced. The study finally learned that miR-181a can reduce the level of MG inflammatory factors by targeting the expression of TRIM9 and has the effect of improving the symptoms of MG.
Collapse
Affiliation(s)
- Qiang Wang
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550002, Guizhou, China
| | - Yunquan Liu
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550002, Guizhou, China
| | - Shixiang Kuang
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550002, Guizhou, China
| | - Ruozhao Li
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550002, Guizhou, China
| | - Ning Weng
- The Second Clinical Medical College of Guizhou University of Traditional Chinese Medicine, Guiyang 550002, Guizhou, China
| | - Zhichao Zhou
- Department of Neurology, Zhuji Affiliated Hospital of Shaoxing University, Zhuji 311800, Zhejiang, China
| |
Collapse
|
14
|
Lee JD, Woodruff TM. The emerging role of complement in neuromuscular disorders. Semin Immunopathol 2021; 43:817-828. [PMID: 34705082 DOI: 10.1007/s00281-021-00895-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
The complement cascade is a key arm of the immune system that protects the host from exogenous and endogenous toxic stimuli through its ability to potently regulate inflammation, phagocytosis, and cell lysis. Due to recent clinical trial successes and drug approvals for complement inhibitors, there is a resurgence in targeting complement as a therapeutic approach to prevent ongoing tissue destruction in several diseases. In particular, neuromuscular diseases are undergoing a recent focus, with demonstrated links between complement activation and disease pathology. This review aims to provide a comprehensive overview of complement activation and its role during the initiation and progression of neuromuscular disorders including myasthenia gravis, amyotrophic lateral sclerosis, and Duchenne muscular dystrophy. We will review the preclinical and clinical evidence for complement in these diseases, with an emphasis on the complement-targeting drugs in clinical trials for these indications.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
15
|
Liu Y, Yang CL, Yang B, Du T, Li XL, Zhang P, Ge MR, Lian Y, Li H, Liu YD, Duan RS. Prophylactic administration of fingolimod (FTY720) ameliorated experimental autoimmune myasthenia gravis by reducing the number of dendritic cells, follicular T helper cells and antibody-secreting cells. Int Immunopharmacol 2021; 96:107511. [PMID: 33915521 DOI: 10.1016/j.intimp.2021.107511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 11/28/2022]
Abstract
Fingolimod (FTY720), a sphingosine 1-phosphate (S1P) receptor antagonist, possesses potent immunomodulatory activity via lymphocyte homing. The effects of FTY720 have been widely studied in various T-cell-mediated autoimmune diseases, while the immunomodulatory effects on experimental autoimmune myasthenia gravis (EAMG), a typical disease model for antibody-mediated autoimmunity, remain elusive. In the present study, FTY720 was administered to EAMG rats as prophylaxis. The clinical scores were recorded every other day, and serum antibodies at different time points were measured by enzyme-linked immunosorbent assay (ELISA). The immune cell subsets in the spleen, bone marrow, circulation, and thymus were determined by flow cytometry. The prophylactic administration alleviated EAMG symptoms by reducing the level of serum antibodies IgG and its isotype IgG2b on days 30 and 46 post immunization, as well as IgG and Ig kappa antibody-secreting cells in the spleen and bone marrow. The mitigated humoral immune response can be attributed to the decreased dendritic cells, follicular T help cells (Tfh) and Tfh subsets (Tfh1, Tfh2, and Tfh17), and T helper cell subsets (Th1, Th2, and Th17) in the spleen. The promotion of lymphocyte homing and inhibition of thymocyte egress contribute to the effects of FTY720 on these effector T cell subsets. Overall, the prophylactic administration of FTY720 ameliorated EAMG partially by regulating humoral immune response,suggesting that FTY720 could be part of a pharmacological strategy for managing myasthenia gravis.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China.
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Bing Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Meng-Ru Ge
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China.
| | - Ying Lian
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Yu-Dong Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China.
| | - Rui-Sheng Duan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China; Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, China; Shandong Institute of Neuroimmunology, China.
| |
Collapse
|
16
|
Huang H, Ran H, Liu X, Yu L, Qiu L, Lin Z, Ou C, Lu Y, Yang W, Liu W. Leflunomide ameliorates experimental autoimmune myasthenia gravis by regulating humoral and cellular immune responses. Int Immunopharmacol 2021; 93:107434. [PMID: 33556668 DOI: 10.1016/j.intimp.2021.107434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/12/2022]
Abstract
Leflunomide, an immunosuppressive disease-modifying anti-rheumatic drug (DMARD), is widely used in the treatment of rheumatoid arthritis (RA), psoriatic arthritis (PA) as well as multiple sclerosis. However, its role in myasthenia gravis (MG) has not yet been clearly explored. Here, we investigated the effect of leflunomide on experimental autoimmune myasthenia gravis (EAMG) in vivo and in vitro. The results demonstrated that leflunomide alleviated the severity of EAMG associated with reduced serum total anti-acetylcholine receptor (AChR) IgG levels. During the development of EAMG, the increase of follicular helper T cells (Tfh) 1, Tfh 17 cells and decrease of follicular regulatory T cells (Tfr) were reversely altered after leflunomide administration. Our work further found that leflunomide might inhibit Tfh cells through the IL-21/STAT3 pathway to reduce the secretion of antibodies by B cells. In addition, leflunomide rebuilt the balance of Th1/Th2/Th17/Treg subsets. These results suggested that leflunomide ameliorated EAMG severity by regulating humoral immune responses and Th cell profiles thereby providing a novel effective treatment strategy for MG.
Collapse
Affiliation(s)
- Huan Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Hao Ran
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoxi Liu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lu Yu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Li Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Zhongqiang Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Changyi Ou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Yaru Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Wenhao Yang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Weibin Liu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
17
|
Mantegazza R, Vanoli F, Frangiamore R, Cavalcante P. Complement Inhibition for the Treatment of Myasthenia Gravis. Immunotargets Ther 2020; 9:317-331. [PMID: 33365280 PMCID: PMC7751298 DOI: 10.2147/itt.s261414] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Generalized myasthenia gravis (gMG) is a rare autoimmune disorder affecting the neuromuscular junction (NMJ). Approximately 80-90% of patients display antibodies directed against the nicotinic acetylcholine receptor (AChR). A major drive of AChR antibody-positive MG pathology is represented by complement activation. The role of the complement cascade has been largely demonstrated in patients and in MG animal models. Complement activation at the NMJ leads to focal lysis of the post-synaptic membrane, disruption of the characteristic folds, and reduction of AChR. Given that the complement system works as an activation cascade, there are many potential targets that can be considered for therapeutic intervention. Preclinical studies have confirmed the efficacy of complement inhibition in ameliorating MG symptoms. Eculizumab, an antibody directed towards C5, has recently been approved for the treatment of AChR antibody-positive gMG. Other complement inhibitors, targeting C5 as well, are currently under phase III study. Complement inhibitors, however, may present prohibitive costs. Therefore, the identification of a subset of patients more or less prone to respond to such therapies would be beneficial. For such purpose, there is a critical need to identify possible biomarkers predictive of therapeutic response, a field not yet sufficiently explored in MG. This review aims to give an overview of the complement cascade involvement in MG, the evolution of complement-inhibiting therapies and possible biomarkers useful to tailor and monitor complement-directed therapies.
Collapse
Affiliation(s)
- Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fiammetta Vanoli
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rita Frangiamore
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paola Cavalcante
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
18
|
Jing F, Huang W, Ma Q, Xu SJ, Wu CJ, Guan YX, Chen B. AEB-071 Ameliorates Muscle Weakness by Altering Helper T Lymphocytes in an Experimental Autoimmune Myasthenia Gravis Rat Model. Med Sci Monit 2020; 26:e924393. [PMID: 32920588 PMCID: PMC7510173 DOI: 10.12659/msm.924393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Myasthenia gravis (MG) is an autoimmune neurological disorder of neuromuscular junctions. In this study we established experimental autoimmune myasthenia gravis (EAMG) rat models to investigate the effects of AEB-071 (AEB), which is a specific inhibitor of protein kinase C that prevents T lymphocyte activation. Material/Methods We utilized animals divided into 4 groups: (1) control rats, (2) EAMG, (3) AEB-071+EAMG, and (4) AZP+EAMG. Drug treatment was continued for 10 days. Ten weeks after immunization we measured body weights, assessed mortality rates, and used Lennon scores to evaluate EAMG grades. We also assessed the proportions of Treg, Th1, Th2, Th17, and lymphocytes using flow cytometry. Results In the absence of drug treatment, we found a significant decline in body weights in the EAMG group in comparison to control rats, and EAMG group rats also had higher Lennon scores (P<0.05). Interestingly, we found that AEB-071 restored the body weight of EAMG rats and the decreased mortality rate compared to AZP treatment. Although a decrease in the number of Treg cells was observed, the proportion of Th lymphocytes was significantly increased in the EAMG group, and AEB-071 treatment decreased the proportion of Th lymphocytes. Conclusions We concluded that AEB-071 treatment imparts beneficial effects in EAMG rat models by reducing mortality rate and restoring Th lymphocyte balance, and thus may be an attractive candidate for use in MG treatment.
Collapse
Affiliation(s)
- Feng Jing
- Department of Neurology, The 8th Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Wei Huang
- Department of Neurology, The 8th Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Qian Ma
- Department of Neurology, The 8th Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Sheng-Jie Xu
- Department of Neurology, The 8th Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| | - Chang-Jin Wu
- Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Yu-Xiu Guan
- Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Bing Chen
- Department of Neurology, The 8th Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China (mainland)
| |
Collapse
|
19
|
Zhao J, Jing J, Zhao W, Li X, Hou L, Zheng C, Kong Q, Li W, Yao X, Chang L, Li H, Mu L, Wang G, Wang J. Osteopontin exacerbates the progression of experimental autoimmune myasthenia gravis by affecting the differentiation of T cell subsets. Int Immunopharmacol 2020; 82:106335. [PMID: 32109680 DOI: 10.1016/j.intimp.2020.106335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 11/23/2022]
Abstract
Osteopontin (OPN) is a multifunctional extracellular matrix phosphoprotein that has a specific and complicated structure, and contributes to numerous physiological and pathological activities. The mechanism of OPN in many diseases has been confirmed; however, the role of OPN in myasthenia gravis (MG) remains unclear. In this study, we recombined rat OPN protein in vitro, and assessed how OPN affects the development of autoimmunity using an experimental autoimmune myasthenia gravis (EAMG) rat model. The results showed that the concentration of OPN in serum was up-regulated. Both mRNA and protein levels in splenocytes increased in the EAMG model. OPN treatment in vitro strongly promoted the differentiation of Th1 cells, and inhibited the differentiation of Treg cells. Intraperitoneal injection of OPN revealed the early incidence of EAMG, and more serious disease. This effect was accompanied by an increased percentage of Th1 cells. In conclusion, OPN likely exacerbates the pathogenesis of EAMG by promoting the differentiation of Th1 cells and inhibiting the differentiation of Treg cells.
Collapse
Affiliation(s)
- Jiarui Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jia Jing
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Xinrong Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Lixuan Hou
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Chunfeng Zheng
- The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar City, Heilongjiang 161000, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Wenjin Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Xiuhua Yao
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Lulu Chang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Ministry of Education Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Ministry of Education Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, Heilongjiang 150086, China.
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China.
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
20
|
Rinaldi E, Consonni A, Cordiglieri C, Sacco G, Crasà C, Fontana A, Morelli L, Elli M, Mantegazza R, Baggi F. Therapeutic Effect of Bifidobacterium Administration on Experimental Autoimmune Myasthenia Gravis in Lewis Rats. Front Immunol 2019; 10:2949. [PMID: 31956324 PMCID: PMC6951413 DOI: 10.3389/fimmu.2019.02949] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/02/2019] [Indexed: 12/13/2022] Open
Abstract
Beneficial effects of probiotics on gut microbiota homeostasis and inflammatory immune responses suggested the investigation of their potential clinical efficacy in experimental models of autoimmune diseases. Indeed, administration of two bifidobacteria and lactobacilli probiotic strains prevented disease manifestations in the Lewis rat model of Myasthenia Gravis (EAMG). Here, we demonstrate the clinical efficacy of therapeutic administration of vital bifidobacteria (i.e., from EAMG onset). The mechanisms involved in immunomodulation were investigated with ex vivo and in vitro experiments. Improvement of EAMG symptoms was associated to decreased anti-rat AChR antibody levels, and differential expression of TGFβ and FoxP3 immunoregulatory transcripts in draining lymph nodes and spleen of treated-EAMG rats. Exposure of rat bone marrow-derived dendritic cells to bifidobacteria or lactobacilli strains upregulated toll-like receptor 2 mRNA expression, a key molecule involved in bacterium recognition via lipotheicoic acid. Live imaging experiments of AChR-specific effector T cells, co-cultured with BMDCs pre-exposed to bifidobacteria, demonstrated increased percentages of motile effector T cells, suggesting a hindered formation of TCR-peptide-MHC complex. Composition of gut microbiota was studied by 16S rRNA gene sequencing, and α and β diversity were determined in probiotic treated EAMG rats, with altered ratios between Tenericutes and Verrucomicrobia (phylum level), and Ruminococcaceae and Lachnospiraceae (family level). Moreover, the relative abundance of Akkermansia genus was found increased compared to healthy and probiotic treated EAMG rats. In conclusion, our findings confirms that the administration of vital bifidobacteria at EAMG onset has beneficial effects on disease progression; this study further supports preclinical research in human MG to evaluate probiotic efficacy as supplementary therapy in MG.
Collapse
Affiliation(s)
- Elena Rinaldi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandra Consonni
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Chiara Cordiglieri
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Grazia Sacco
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Camilla Crasà
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandra Fontana
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Lorenzo Morelli
- Department for Sustainable Food Process, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Marina Elli
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, Italy
| | - Renato Mantegazza
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fulvio Baggi
- Neurology IV - Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
21
|
Song J, Xi JY, Yu WB, Yan C, Luo SS, Zhou L, Zhu WH, Lu JH, Dong Q, Xiao BG, Zhao CB. Inhibition of ROCK activity regulates the balance of Th1, Th17 and Treg cells in myasthenia gravis. Clin Immunol 2019; 203:142-153. [PMID: 31078707 DOI: 10.1016/j.clim.2019.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 01/10/2023]
Abstract
Aberrant ROCK activation has been found in patients with several autoimmune diseases, but the role of ROCK in myasthenia gravis (MG) has not yet been clearly investigated. Here, we demonstrated that ROCK activity was significantly higher in peripheral blood mononuclear cells (PBMCs) from MG patients. ROCK inhibitor Fasudil down-regulated the proportions of Th1 and Th17 cells in PBMCs of MG patients in vitro. Intraperitoneal injection of Fasudil ameliorated the severity of experimental autoimmune myasthenia gravis (EAMG) rats and restored the balance of Th1/Th2/Th17/Treg subsets. Furthermore, Fasudil inhibited the proliferation of antigen-specific Th1 and Th17 cells, and inhibited CD4 + T cells differentiated into Th1 and Th17 through decreasing phosphorylated Stat1 and Stat3, but promoted Treg cell differentiation through increasing phosphorylated Stat5. We conclude that dysregulated ROCK activity may be involved in the pathogenic immune response of MG and inhibition of ROCK activity might serve as a novel treatment strategy for MG.
Collapse
Affiliation(s)
- Jie Song
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Jian-Ying Xi
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Wen-Bo Yu
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Su-Shan Luo
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Lei Zhou
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Wen-Hua Zhu
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Jia-Hong Lu
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Bao-Guo Xiao
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China
| | - Chong-Bo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, 12 Middle Wulumuqi Road, Shanghai 200040, China.
| |
Collapse
|
22
|
Song J, Lei X, Jiao W, Song Y, Chen W, Li J, Chen Z. Effect of Qiangji Jianli decoction on mitochondrial respiratory chain activity and expression of mitochondrial fusion and fission proteins in myasthenia gravis rats. Sci Rep 2018; 8:8623. [PMID: 29872094 PMCID: PMC5988663 DOI: 10.1038/s41598-018-26918-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023] Open
Abstract
Myasthenia gravis (MG) is an autoimmune neuromuscular disease characterized by the production of antibodies against acetylcholine receptors (AChRs). Qiangji Jianli (QJJL) decoction is an effective traditional Chinese medicine (TCM) that is used to treat MG. Our study aimed to investigate the effect of QJJL decoction on MG and to clarify the mechanism by which QJJL regulates mitochondrial energy metabolism and mitochondrial fusion and fission (MFF). SPF female Lewis rats were administered Rat 97–116 peptides to induce experimental autoimmune myasthenia gravis (EAMG). The treatment groups received QJJL decoction (7.8 g/kg, 15.6 g/kg and 23.4 g/kg). Mitochondria were extracted from gastrocnemius tissue samples to detect respiratory chain complex enzymatic activity. Quantitative PCR and western blot analysis were performed to detect Mfn1/2, Opa1, Drp1 and Fis1 mRNA and protein expression, respectively, in the mitochondria. Transmission electron microscopy examination was performed to show the improvement of mitochondria and myofibrils after QJJL treatment. The results indicated that QJJL decoction may attenuate MG by promoting the enzymatic activity of respiratory chain complexes to improve energy metabolism. Moreover, QJJL decoction increased Mfn1/2, Opa1, Drp1 and Fis1 mRNA and protein expression to exert its curative effect on MFF. Thus, QJJL decoction may be a promising therapy for MG.
Collapse
Affiliation(s)
- Jingwei Song
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaowen Lei
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wei Jiao
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yafang Song
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Weijing Chen
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jinqiu Li
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhiwei Chen
- Institute of Spleen-Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| |
Collapse
|
23
|
Zhang P, Yang CL, Liu RT, Li H, Zhang M, Zhang N, Yue LT, Wang CC, Dou YC, Duan RS. Toll-like receptor 9 antagonist suppresses humoral immunity in experimental autoimmune myasthenia gravis. Mol Immunol 2018; 94:200-208. [PMID: 29331804 DOI: 10.1016/j.molimm.2018.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/04/2018] [Accepted: 01/08/2018] [Indexed: 01/28/2023]
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.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Chun-Lin Yang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Ru-Tao Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Heng Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Min Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Na Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Long-Tao Yue
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Cong-Cong Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Ying-Chun Dou
- College of Basic Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China.
| |
Collapse
|
24
|
Yang G, Yang X, Zhang J, Li G, Zheng D, Peng A, Hu J, Xu L, Yang B, Yang H, Zhou W, Tuzun E, Li J. Transcriptional repressor Blimp1 regulates follicular regulatory T-cell homeostasis and function. Immunology 2017; 153:105-117. [PMID: 28833081 DOI: 10.1111/imm.12815] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023] Open
Abstract
The B-lymphocyte-induced maturation protein 1 (Blimp1) regulates T-cell homeostasis and function. Loss of Blimp1 could double the proportion of follicular regulatory T (Tfr) cells. However, the effects that Blimp1 may have on the function of Tfr cells remain unknown. Here we document the function for Blimp1 in Tfr cells in vitro and in vivo. Data presented in this study demonstrate that Tfr cells indirectly inhibit the activation and differentiation of B cells by negatively regulating follicular helper T cells, so lowering the secretion of antibody. Lack of Blimp1 makes the immune suppression function of Tfr cells impaired in vitro. In the in vivo study, adoptive transfer of Tfr cells could reduce immune responses in germinal centres and relieve the muscle weakness symptoms of mice with experimental autoimmune myasthenia gravis. Blimp1 deficiency resulted in reduced suppressive ability of Tfr cells. This study identifies that Tfr cells are potent suppressors of immunity and are controlled by Blimp1.
Collapse
Affiliation(s)
- Guang Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaosu Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Junmei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Guancheng Li
- Cancer Research Institute, Central South University, Changsha, China
| | - Dandan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Anjiao Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jue Hu
- Department of Neurology, Changsha Central Hospital, Changsha, China
| | - Liqun Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Baifeng Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenbin Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Erdem Tuzun
- Department of Neurology, University of Istanbul, Istanbul, Turkey
| | - Jing Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Consonni A, Sharma S, Schön K, Lebrero-Fernández C, Rinaldi E, Lycke NY, Baggi F. A Novel Approach to Reinstating Tolerance in Experimental Autoimmune Myasthenia Gravis Using a Targeted Fusion Protein, mCTA1-T146. Front Immunol 2017; 8:1133. [PMID: 28959261 PMCID: PMC5604076 DOI: 10.3389/fimmu.2017.01133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Reinstating tissue-specific tolerance has attracted much attention as a means to treat autoimmune diseases. However, despite promising results in rodent models of autoimmune diseases, no established tolerogenic therapy is clinically available yet. In the experimental autoimmune myasthenia gravis (EAMG) model several protocols have been reported that induce tolerance against the prime disease-associated antigen, the acetylcholine receptor (AChR) at the neuromuscular junction. Using the whole AChR, the extracellular part or peptides derived from the receptor, investigators have reported variable success with their treatments, though, usually relatively large amounts of antigen has been required. Hence, there is a need for better formulations and strategies to improve on the efficacy of the tolerance-inducing therapies. Here, we report on a novel targeted fusion protein carrying the immunodominant peptide from AChR, mCTA1–T146, which given intranasally in repeated microgram doses strongly suppressed induction as well as ongoing EAMG disease in mice. The results corroborate our previous findings, using the same fusion protein approach, in the collagen-induced arthritis model showing dramatic suppressive effects on Th1 and Th17 autoaggressive CD4 T cells and upregulated regulatory T cell activities with enhanced IL10 production. A suppressive gene signature with upregulated expression of mRNA for TGFβ, IL10, IL27, and Foxp3 was clearly detectable in lymph node and spleen following intranasal treatment with mCTA1–T146. Amelioration of EAMG disease was accompanied by reduced loss of muscle AChR and lower levels of anti-AChR serum antibodies. We believe this targeted highly effective fusion protein mCTA1–T146 is a promising candidate for clinical evaluation in myasthenia gravis patients.
Collapse
Affiliation(s)
- Alessandra Consonni
- Neurology IV, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sapna Sharma
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernández
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elena Rinaldi
- Neurology IV, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| | - Nils Yngve Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Fulvio Baggi
- Neurology IV, Fondazione I.R.C.C.S. Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
26
|
ONX-0914, a selective inhibitor of immunoproteasome, ameliorates experimental autoimmune myasthenia gravis by modulating humoral response. J Neuroimmunol 2017; 311:71-78. [PMID: 28844501 DOI: 10.1016/j.jneuroim.2017.08.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/29/2017] [Accepted: 08/18/2017] [Indexed: 11/23/2022]
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.
Collapse
|
27
|
Caspase-1 inhibitor regulates humoral responses in experimental autoimmune myasthenia gravis via IL-6- dependent inhibiton of STAT3. Neurosci Lett 2017; 656:169-176. [DOI: 10.1016/j.neulet.2017.05.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 04/12/2017] [Accepted: 05/19/2017] [Indexed: 12/13/2022]
|
28
|
Rapamycin alleviates inflammation and muscle weakness, while altering the Treg/Th17 balance in a rat model of myasthenia gravis. Biosci Rep 2017; 37:BSR20170767. [PMID: 28655853 PMCID: PMC5518538 DOI: 10.1042/bsr20170767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/25/2017] [Accepted: 06/26/2017] [Indexed: 12/19/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease commonly treated with immunosuppressants. We evaluated the novel immunosuppressant, rapamycin (RAPA), in a rat model of experimental autoimmune MG (EAMG). Mortality rates in the RAPA (12%) were significantly down compared with the EAMG (88%) or cyclophosphamide (CTX) (68%) intervention groups. Muscular weakness decreased after both RAPA and CTX treatment. However, Lennon scores were lower (1.74 ± 0.49, 3.39 ± 0.21, and 3.81 ± 0.22 in RAPA, CTX, and EAMG groups, respectively), and body weights (203.12 ± 4.13 g, 179.23 ± 2.13 g, and 180.13 ± 5.13 g in RAPA, CTX, and EAMG groups, respectively) were significantly higher, only in the RAPA group. The proportion of regulatory T cells (Treg) significantly increased, while that of Th17 cells significantly decreased in the RAPA group compared with the EAMG group. In comparison, CTX intervention resulted in increased Th17 but significantly decreased Tregs. Hence, RAPA can be more effectively used in comparison with CTX to treat MG, with an efficacy higher than that of CTX. In addition, our results suggest RAPA’s efficacy in alleviating symptoms of MG stems from its ability to correct the Treg/Th17 imbalance observed in MG.
Collapse
|
29
|
Lazaridis K, Baltatzidi V, Trakas N, Koutroumpi E, Karandreas N, Tzartos SJ. Characterization of a reproducible rat EAMG model induced with various human acetylcholine receptor domains. J Neuroimmunol 2017; 303:13-21. [DOI: 10.1016/j.jneuroim.2016.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/04/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023]
|
30
|
Chen W, Meng QF, Sui JK, Wang YJ, Li XL, Liu S, Li H, Wang CC, Li CH, Li YB. Ginsenoside Rb1: The new treatment measure of myasthenia gravis. Int Immunopharmacol 2016; 41:136-143. [DOI: 10.1016/j.intimp.2016.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/06/2016] [Accepted: 08/24/2016] [Indexed: 02/08/2023]
|
31
|
Astilbin ameliorates experimental autoimmune myasthenia gravis by decreased Th17 cytokines and up-regulated T regulatory cells. J Neuroimmunol 2016; 298:138-45. [PMID: 27609287 DOI: 10.1016/j.jneuroim.2016.07.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/16/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
Abstract
Astilbin, a major bioactive compound extracted from Rhizoma smilacis glabrae (RSG), has been reported to possess immunosuppressive properties. Our study first evaluated the effect of astilbin on experimental autoimmune myasthenia gravis (EAMG) in Lewis rats. The results showed that astilbin could attenuate the severity of EAMG by decreasing antigen-specific autoantibodies with up-regulation of regulatory T cells and down-regulation of Th17 cells. In addition to, astilbin also reduced the efficiency of the antigen presenting cells on which the expression of MHC class II decreased. These results suggest that astilbin might be a candidate drug for immunoregulation of EAMG, and provide us new treatment ideas for human myasthenia gravis (MG).
Collapse
|
32
|
Effect of Polygoni Cuspidati Rhizoma et Radix and Its Ingredient Resveratrol on Experimental Autoimmune Myasthenia Gravis by Suppressing Immune Response. CHINESE HERBAL MEDICINES 2016. [DOI: 10.1016/s1674-6384(16)60047-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
33
|
Li H, Zhang M, Wang CC, Li XL, Zhang P, Yue LT, Miao S, Dou YC, Li YB, Duan RS. ROCK inhibitor abolishes the antibody response in experimental autoimmune myasthenia gravis. Mol Cell Neurosci 2016; 74:106-13. [DOI: 10.1016/j.mcn.2016.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 04/18/2016] [Accepted: 05/06/2016] [Indexed: 02/08/2023] Open
|
34
|
Mantegazza R, Cordiglieri C, Consonni A, Baggi F. Animal models of myasthenia gravis: utility and limitations. Int J Gen Med 2016; 9:53-64. [PMID: 27019601 PMCID: PMC4786081 DOI: 10.2147/ijgm.s88552] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Myasthenia gravis (MG) is a chronic autoimmune disease caused by the immune attack of the neuromuscular junction. Antibodies directed against the acetylcholine receptor (AChR) induce receptor degradation, complement cascade activation, and postsynaptic membrane destruction, resulting in functional reduction in AChR availability. Besides anti-AChR antibodies, other autoantibodies are known to play pathogenic roles in MG. The experimental autoimmune MG (EAMG) models have been of great help over the years in understanding the pathophysiological role of specific autoantibodies and T helper lymphocytes and in suggesting new therapies for prevention and modulation of the ongoing disease. EAMG can be induced in mice and rats of susceptible strains that show clinical symptoms mimicking the human disease. EAMG models are helpful for studying both the muscle and the immune compartments to evaluate new treatment perspectives. In this review, we concentrate on recent findings on EAMG models, focusing on their utility and limitations.
Collapse
Affiliation(s)
- Renato Mantegazza
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Chiara Cordiglieri
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Alessandra Consonni
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| | - Fulvio Baggi
- Neurology IV Unit, Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute "Carlo Besta", Milan, Italy
| |
Collapse
|
35
|
Exosomes derived from atorvastatin-modified bone marrow dendritic cells ameliorate experimental autoimmune myasthenia gravis by up-regulated levels of IDO/Treg and partly dependent on FasL/Fas pathway. J Neuroinflammation 2016; 13:8. [PMID: 26757900 PMCID: PMC4710023 DOI: 10.1186/s12974-016-0475-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 01/03/2016] [Indexed: 12/20/2022] Open
Abstract
Background Previously, we have demonstrated that spleen-derived dendritic cells (DCs) modified with atorvastatin suppressed immune responses of experimental autoimmune myasthenia gravis (EAMG). However, the effects of exosomes derived from atorvastatin-modified bone marrow DCs (BMDCs) (statin-Dex) on EAMG are still unknown. Methods Immunophenotypical characterization of exosomes from atorvastatin- and dimethylsulfoxide (DMSO)-modified BMDCs was performed by electron microscopy, flow cytometry, and western blotting. In order to investigate whether statin-DCs-derived exosomes (Dex) could induce immune tolerance in EAMG, we administrated statin-Dex, control-Dex, or phosphate-buffered saline (PBS) into EAMG rats via tail vein injection. The tracking of injected Dex and the effect of statin-Dex injection on endogenous DCs were performed by immunofluorescence and flow cytometry, respectively. The number of Foxp3+ cells in thymuses was examined using immunocytochemistry. Treg cells, cytokine secretion, lymphocyte proliferation, cell viability and apoptosis, and the levels of autoantibody were also carried out to evaluate the effect of statin-Dex on EAMG rats. To further investigate the involvement of FasL/Fas in statin-Dex-induced apoptosis, the underlying mechanisms were studied by FasL neutralization assays. Results Our data showed that the systemic injection of statin-Dex suppressed the clinical symptoms of EAMG rats. These statin-Dex had immune regulation functions in immune organs, such as the spleen, thymus, and popliteal and inguinal lymph nodes. Furthermore, statin-Dex exerted their immunomodulatory effects in vivo by decreasing the expression of CD80, CD86, and MHC class II on endogenous DCs. Importantly, the therapeutic effects of statin-Dex on EAMG rats were associated with up-regulated levels of indoleamine 2,3-dioxygenase (IDO)/Treg and partly dependent on FasL/Fas pathway, which finally resulted in decreased synthesis of anti-R97–116 IgG, IgG2a, and IgG2b antibodies. Conclusions Our data suggest that atorvastatin-induced immature BMDCs are able to secrete tolerogenic Dex, which are involved in the suppression of immune responses in EAMG rats. Importantly, our study provides a novel cell-free approach for the treatment of autoimmune diseases.
Collapse
|
36
|
Li H, Wang CC, Zhang M, Li XL, Zhang P, Yue LT, Miao S, Wang S, Liu Y, Li YB, Duan RS. Statin-modified dendritic cells regulate humoral immunity in experimental autoimmune myasthenia gravis. Mol Cell Neurosci 2015; 68:284-92. [DOI: 10.1016/j.mcn.2015.08.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/03/2015] [Accepted: 08/19/2015] [Indexed: 12/16/2022] Open
|
37
|
Wang CC, Li H, Zhang M, Li XL, Yue LT, Zhang P, Zhao Y, Wang S, Duan RN, Li YB, Duan RS. Caspase-1 inhibitor ameliorates experimental autoimmune myasthenia gravis by innate dendric cell IL-1-IL-17 pathway. J Neuroinflammation 2015; 12:118. [PMID: 26071315 PMCID: PMC4470006 DOI: 10.1186/s12974-015-0334-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/30/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND IL-1β has been shown to play a pivotal role in autoimmunity. Cysteinyl aspartate-specific proteinase-1 (caspase-1) inhibitor may be an important drug target for autoimmune diseases. However, the effects of caspase-1 inhibitor on myasthenia gravis (MG) remain undefined. METHODS To investigate the effects of caspase-1 inhibitor on experimental autoimmune myasthenia gravis (EAMG), an animal model of MG, caspase-1 inhibitor was administered to Lewis rats immunized with region 97-116 of the rat AChR α subunit (R97-116 peptide) in complete Freund's adjuvant. The immunophenotypical characterization by flow cytometry and the levels of autoantibody by ELISA were carried out to evaluate the neuroprotective effect of caspase-1 inhibitor. RESULTS We found that caspase-1 inhibitor improved EAMG clinical symptom, which was associated with decreased IL-17 production by CD4+ T cells and γδ T cells, lower affinity of anti-R97-116 peptide IgG. Caspase-1 inhibitor decreased expression of CD80, CD86, and MHC class II on DCs, as well as intracellular IL-1β production from DCs. In addition, caspase-1 inhibitor treatment inhibited R97-116 peptide-specific cell proliferation and decreased follicular helper T cells relating to EAMG development. CONCLUSIONS Our results suggest that caspase-1 inhibitor ameliorates experimental autoimmune myasthenia gravis by innate DC IL-1-IL-17 pathway and provides new evidence that caspase-1 is an important drug target in the treatment of MG and other autoimmune diseases.
Collapse
Affiliation(s)
- Cong-Cong Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Heng Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Min Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Xiao-Li Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Long-Tao Yue
- Central Laboratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Peng Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Yue Zhao
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Shan Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Ruo-Nan Duan
- School of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Yan-Bin Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, China.
| |
Collapse
|
38
|
Xu H, Zhang M, Li XL, Li H, Yue LT, Zhang XX, Wang CC, Wang S, Duan RS. Low and high doses of ursolic acid ameliorate experimental autoimmune myasthenia gravis through different pathways. J Neuroimmunol 2015; 281:61-7. [PMID: 25867469 DOI: 10.1016/j.jneuroim.2015.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 01/08/2015] [Accepted: 02/28/2015] [Indexed: 10/23/2022]
Abstract
Myasthenia gravis (MG) is an autoimmune disease characterized by fatigable muscle weakness. Ursolic acid (UA) is a pentacyclic triterpenoid with anti-inflammatory and immunomodulatory properties, especially inhibiting IL-17. We found that UA ameliorated the symptoms of experimental autoimmune myasthenia gravis (EAMG), a rat model of MG. Although both the low and high doses of UA shifted Th17 to Th2 cytokines, other mechanisms were dose dependent. The low dose enhanced Fas-mediated apoptosis, whereas the high dose up-regulated Treg cells and reduced the concentrations of IgG2b antibodies. These findings suggest a new strategy to treat EAMG and even human MG.
Collapse
Affiliation(s)
- Hua Xu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China; Department of Neurology, Taian City Central Hospital, Taian 271000, PR China
| | - Min Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Xiao-Li Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Heng Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Long-Tao Yue
- Central Laboratory, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Xin-Xin Zhang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Cong-Cong Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Shan Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China.
| |
Collapse
|
39
|
Losen M, Martinez-Martinez P, Molenaar PC, Lazaridis K, Tzartos S, Brenner T, Duan RS, Luo J, Lindstrom J, Kusner L. Standardization of the experimental autoimmune myasthenia gravis (EAMG) model by immunization of rats with Torpedo californica acetylcholine receptors--Recommendations for methods and experimental designs. Exp Neurol 2015; 270:18-28. [PMID: 25796590 PMCID: PMC4466156 DOI: 10.1016/j.expneurol.2015.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 12/21/2022]
Abstract
Myasthenia gravis (MG) with antibodies against the acetylcholine receptor (AChR) is characterized by a chronic, fatigable weakness of voluntary muscles. The production of autoantibodies involves the dysregulation of T cells which provide the environment for the development of autoreactive B cells. The symptoms are caused by destruction of the postsynaptic membrane and degradation of the AChR by IgG autoantibodies, predominantly of the G1 and G3 subclasses. Active immunization of animals with AChR from mammalian muscles, AChR from Torpedo or Electrophorus electric organs, and recombinant or synthetic AChR fragments generates a chronic model of MG, termed experimental autoimmune myasthenia gravis (EAMG). This model covers cellular mechanisms involved in the immune response against the AChR, e.g. antigen presentation, T cell-help and regulation, B cell selection and differentiation into plasma cells. Our aim is to define standard operation procedures and recommendations for the rat EAMG model using purified AChR from the Torpedo californica electric organ, in order to facilitate more rapid translation of preclinical proof of concept or efficacy studies into clinical trials and, ultimately, clinical practice.
Collapse
Affiliation(s)
- Mario Losen
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| | - Pilar Martinez-Martinez
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Peter C Molenaar
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | | - Socrates Tzartos
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, PR China
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Jon Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Linda Kusner
- Department of Pharmacology & Physiology, The George Washington University, Washington, DC, USA
| |
Collapse
|
40
|
Zhang J, Jia G, Liu Q, Hu J, Yan M, Yang B, Yang H, Zhou W, Li J. Silencing miR-146a influences B cells and ameliorates experimental autoimmune myasthenia gravis. Immunology 2015; 144:56-67. [PMID: 24962817 DOI: 10.1111/imm.12347] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/30/2014] [Accepted: 06/17/2014] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs have been shown to be important regulators of immune homeostasis as patients with aberrant microRNA expression appeared to be more susceptible to autoimmune diseases. We recently found that miR-146a was up-regulated in activated B cells in response to rat acetylcholine receptor (AChR) α-subunit 97-116 peptide, and this up-regulation was significantly attenuated by AntagomiR-146a. Our data also demonstrated that silencing miR-146a with its inhibitor AntagomiR-146a effectively ameliorated clinical myasthenic symptoms in mice with ongoing experimental autoimmune myasthenia gravis. Furthermore, multiple defects were observed after miR-146a was knocked down in B cells, including decreased anti-R97-116 antibody production and class switching, reduced numbers of plasma cells, memory B cells and B-1 cells, and weakened activation of B cells. Previously, miR-146a has been identified as a nuclear factor-κB-dependent gene and predicted to base pair with the tumour necrosis factor receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 1 (IRAK1) genes to regulate the immune response. However, our study proved that miR-146a inhibition had no effect on the expression of TRAF6 and IRAK1 in B cells. This result suggests that the function of miR-146a in B cells does not involve these two target molecules. We conclude that silencing miR-146a exerts its therapeutic effects by influencing the B-cell functions that contribute to the autoimmune pathogenesis of myasthenia gravis.
Collapse
Affiliation(s)
- JunMei Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Deficits in endogenous adenosine formation by ecto-5'-nucleotidase/CD73 impair neuromuscular transmission and immune competence in experimental autoimmune myasthenia gravis. Mediators Inflamm 2015; 2015:460610. [PMID: 25691808 PMCID: PMC4322825 DOI: 10.1155/2015/460610] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/20/2014] [Accepted: 08/21/2014] [Indexed: 11/18/2022] Open
Abstract
AMP dephosphorylation via ecto-5'-nucleotidase/CD73 is the rate limiting step to generate extracellular adenosine (ADO) from released adenine nucleotides. ADO, via A2A receptors (A2ARs), is a potent modulator of neuromuscular and immunological responses. The pivotal role of ecto-5'-nucleotidase/CD73, in controlling extracellular ADO formation, prompted us to investigate its role in a rat model of experimental autoimmune myasthenia gravis (EAMG). Results show that CD4(+)CD25(+)FoxP3(+) regulatory T cells express lower amounts of ecto-5'-nucleotidase/CD73 as compared to controls. Reduction of endogenous ADO formation might explain why proliferation of CD4(+) T cells failed upon blocking A2A receptors activation with ZM241385 or adenosine deaminase in EAMG animals. Deficits in ADO also contribute to neuromuscular transmission failure in EAMG rats. Rehabilitation of A2AR-mediated immune suppression and facilitation of transmitter release were observed by incubating the cells with the nucleoside precursor, AMP. These findings, together with the characteristic increase in serum adenosine deaminase activity of MG patients, strengthen our hypothesis that the adenosinergic pathway may be dysfunctional in EAMG. Given that endogenous ADO formation is balanced by ecto-5'-nucleotidase/CD73 activity and that A2ARs exert a dual role to restore use-dependent neurocompetence and immune suppression in myasthenics, we hypothesize that stimulation of the two mechanisms may have therapeutic potential in MG.
Collapse
|
42
|
Yao X, Kong Q, Xie X, Wang J, Li N, Liu Y, Sun B, Li Y, Wang G, Li W, Qu S, Zhao H, Wang D, Liu X, Zhang Y, Mu L, Li H. Neutralization of interleukin-9 ameliorates symptoms of experimental autoimmune myasthenia gravis in rats by decreasing effector T cells and altering humoral responses. Immunology 2014; 143:396-405. [PMID: 24850614 DOI: 10.1111/imm.12322] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 12/21/2022] Open
Abstract
Interleukin-9 (IL-9) was initially thought to be a type 2 T helper (Th2)-associated cytokine involved in the regulation of autoimmune responses by affecting multiple cell types. However, it was recently shown that IL-9-producing CD4+ T cells represent a discrete subset of Th cells, designated Th9 cells. Although Th9 cells have been shown to be important in many diseases, their roles in myasthenia gravis (MG) are unclear. The aim of this study was to determine whether IL-9 and Th9 cells promote the progression of experimental autoimmune myasthenia gravis (EAMG). The results showed that the percentage of Th9 cells changed during the progression of EAMG, accompanied by an up-regulation of IL-9. Blocking IL-9 activity with antibodies against IL-9 inhibited EAMG-associated pathology in rats and reduced serum anti-acetylcholine receptor IgG levels. Neutralization of IL-9 altered the Th subset distribution in EAMG, reducing the number of Th1 cells and increasing the number of regulatory T cells. Administration of an anti-IL-9 antibody may represent an effective therapeutic strategy for MG-associated pathologies or other T-cell- or B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Xiuhua Yao
- Department of Neurobiology, Provincial Key Laboratory of Neurobiology, Harbin Medical University, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Li N, Wang G, Yao X, Kong Q, Shang X, Xie X, Wang J, Kang X, Jin L, Wang G, Li H, Mu L, Sun B. Adenosine receptor expression in a rat model of experimental autoimmune myasthenia gravis. Cell Immunol 2014; 290:217-25. [PMID: 25086239 DOI: 10.1016/j.cellimm.2014.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/05/2014] [Accepted: 07/14/2014] [Indexed: 01/25/2023]
Abstract
Extracellular adenosine is an essential negative regulator of immune reactions that acts by signaling via 4 distinct adenosine receptors. We evaluated adenosine receptor expression in Lewis rats presenting with experimental autoimmune myasthenia gravis (EAMG) to determine whether the expression of adenosine receptors are changed in the development and progression of EAMG. Lymphocyte A1AR and A2AAR mRNA and protein levels from lymphocytes harvested from the lymph nodes, spleen, and peripheral blood mononuclear cells (PBMCs) of EAMG rats were decreased. A modest but not significant increase in A2BAR levels was observed in EAMG lymphocytes harvested from lymph nodes and PBMCs. No changes in A3AR expression were observed in lymphocytes harvested from lymph nodes, spleen, or PBMCs following EAMG induction. Results presented in this report showed that the expression levels and the distribution pattern of adenosine receptors were altered in EAMG lymphocytes.
Collapse
Affiliation(s)
- Na Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China; Department of Histology and Embryology, Harbin Medical University, Heilongjiang 150081, China
| | - Geng Wang
- Department of Anatomy, Harbin Medical University, Heilongjiang 150081, China
| | - Xiuhua Yao
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Xiaoyu Shang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Xiaoli Xie
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Xiaoying Kang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Lianhong Jin
- Department of Histology and Embryology, Harbin Medical University, Heilongjiang 150081, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China.
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang 150081, China.
| |
Collapse
|
44
|
Cordiglieri C, Marolda R, Franzi S, Cappelletti C, Giardina C, Motta T, Baggi F, Bernasconi P, Mantegazza R, Cavalcante P. Innate immunity in myasthenia gravis thymus: pathogenic effects of Toll-like receptor 4 signaling on autoimmunity. J Autoimmun 2014; 52:74-89. [PMID: 24397961 DOI: 10.1016/j.jaut.2013.12.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/15/2013] [Indexed: 12/21/2022]
Abstract
The thymus is the main site of immune sensitization to AChR in myasthenia gravis (MG). In our previous studies we demonstrated that Toll-like receptor (TLR) 4 is over-expressed in MG thymuses, suggesting its involvement in altering the thymic microenvironment and favoring autosensitization and autoimmunity maintenance processes, via an effect on local chemokine/cytokine network. Here, we investigated whether TLR4 signaling may favor abnormal cell recruitment in MG thymus via CCL17 and CCL22, two chemokines known to dictate immune cell trafficking in inflamed organs by binding CCR4. We also investigated whether TLR4 activation may contribute to immunodysregulation, via the production of Th17-related cytokines, known to alter effector T cell (Teff)/regulatory T cell (Treg) balance. We found that CCL17, CCL22 and CCR4 were expressed at higher levels in MG compared to normal thymuses. The two chemokines were mainly detected around medullary Hassall's corpuscles (HCs), co-localizing with TLR4(+) thymic epithelial cells (TECs) and CCR4(+) dendritic cells (DCs), that were present in higher number in MG thymuses compared to controls. TLR4 stimulation in MG TECs increased CCL17 and CCL22 expression and induced the production of Th17-related cytokines. Then, to study the effect of TLR4-stimulated TECs on immune cell interactions and Teff activation, we generated an in-vitro imaging model by co-culturing CD4(+) Th1/Th17 AChR-specific T cells, naïve CD4(+)CD25(+) Tregs, DCs and TECs from Lewis rats. We observed that TLR4 stimulation led to a more pronounced Teff activatory status, suggesting that TLR4 signaling in MG thymic milieu may affect cell-to-cell interactions, favoring autoreactive T-cell activation. Altogether our findings suggest a role for TLR4 signaling in driving DC recruitment in MG thymus via CCL17 and CCL22, and in generating an inflammatory response that might compromise Treg function, favoring autoreactive T-cell pathogenic responses.
Collapse
Affiliation(s)
- Chiara Cordiglieri
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Roberta Marolda
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Sara Franzi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Cristina Cappelletti
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Carmelo Giardina
- Department of Pathological Anatomy, Azienda Ospedaliera Bolognini Seriate, Via Paterno 21, 24068 Seriate Bergamo, Italy.
| | - Teresio Motta
- Department of Pathological Anatomy, Azienda Ospedaliera Bolognini Seriate, Via Paterno 21, 24068 Seriate Bergamo, Italy.
| | - Fulvio Baggi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Pia Bernasconi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Renato Mantegazza
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Paola Cavalcante
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| |
Collapse
|
45
|
Wang J, Zheng S, Xin N, Dou C, Fu L, Zhang X, Chen J, Zhang Y, Geng D, Xiao C, Cui G, Shen X, Lu Y, Wang J, Dong R, Qiao Y, Zhang Y. Identification of Novel MicroRNA Signatures Linked to Experimental Autoimmune Myasthenia Gravis Pathogenesis: Down-Regulated miR-145 Promotes Pathogenetic Th17 Cell Response. J Neuroimmune Pharmacol 2013; 8:1287-302. [DOI: 10.1007/s11481-013-9498-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022]
|
46
|
Xie X, Mu L, Yao X, Li N, Sun B, Li Y, Zhan X, Wang X, Kang X, Wang J, Liu Y, Zhang Y, Wang G, Wang D, Liu X, Kong Q, Li H. ATRA alters humoral responses associated with amelioration of EAMG symptoms by balancing Tfh/Tfr helper cell profiles. Clin Immunol 2013; 148:162-76. [DOI: 10.1016/j.clim.2013.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 01/10/2023]
|
47
|
Marolda R, Ruocco C, Cordiglieri C, Toscani C, Antozzi C, Mantegazza R, Baggi F. Differential targeting of immune-cells by Pixantrone in experimental myasthenia gravis. J Neuroimmunol 2013; 258:41-50. [DOI: 10.1016/j.jneuroim.2013.02.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/22/2013] [Accepted: 02/26/2013] [Indexed: 11/28/2022]
|
48
|
Li XL, Liu Y, Cao LL, Li H, Yue LT, Wang S, Zhang M, Li XH, Dou YC, Duan RS. Atorvastatin-modified dendritic cells in vitro ameliorate experimental autoimmune myasthenia gravis by up-regulated Treg cells and shifted Th1/Th17 to Th2 cytokines. Mol Cell Neurosci 2013; 56:85-95. [PMID: 23541702 DOI: 10.1016/j.mcn.2013.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 01/25/2013] [Accepted: 03/11/2013] [Indexed: 10/27/2022] Open
Abstract
Conventional therapies for autoimmune diseases produce nonspecific immune suppression, which are usually continued lifelong to maintain disease control, and associated with a variety of adverse effects. In this study, we found that spleen-derived dendritic cells (DCs) from the ongoing experimental autoimmune myasthenia gravis (EAMG) rats can be induced into tolerogenic DCs by atorvastatin in vitro. Administration of these tolerogenic DCs to EAMG rats on days 5 and 13 post immunization (p.i.) resulted in improved clinical symptoms, which were associated with increased numbers of CD4(+)CD25(+) T regulatory (Treg) cells and Foxp3 expression, decreased lymphocyte proliferation among lymph node mononuclear cells (MNC), shifted cytokine profile from Th1/Th17 to Th2 type cytokines, decreased level of anti-R97-116 peptide (region 97-116 of the rat acetylcholine receptor α subunit) IgG antibody in serum. These tolerogenic DCs can migrate to spleen, thymus, popliteal and inguinal lymph nodes after they were injected into the EAMG rats intraperitoneally. Furthermore, these tolerogenic DCs played their immunomodulatory effects in vivo mainly by decreased expression of CD86 and MHC class II on endogenous DCs. All these data provided us a new strategy to treat EAMG and even human myasthenia gravis (MG).
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Tüzün E, Allman W, Ulusoy C, Yang H, Christadoss P. Novel animal models of acetylcholine receptor antibody-related myasthenia gravis. Ann N Y Acad Sci 2012; 1274:133-9. [DOI: 10.1111/j.1749-6632.2012.06773.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Li N, Mu L, Wang J, Zhang J, Xie X, Kong Q, Tang W, Yao X, Liu Y, Wang L, Wang G, Wang D, Jin L, Sun B, Li H. Activation of the adenosine A2A receptor attenuates experimental autoimmune myasthenia gravis severity. Eur J Immunol 2012; 42:1140-51. [PMID: 22539289 DOI: 10.1002/eji.201142088] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The adenosine A2A receptor (A2AR) is the major cellular adenosine receptor commonly associated with immunosuppression. Here, we investigated whether A2AR activation holds the potential for impacting the severity of experimental autoimmune myasthenia gravis (EAMG) induced following immunization of Lewis rats with the acetylcholine receptor (AChR) R97-116 peptide. This report demonstrates reduced A2AR expression by both T cells and B cells residing in spleen and lymph nodes following EAMG induction. A2AR stimulation inhibited anti-AChR antibody production and proliferation of AChR-specific lymphocytes in vitro. Inhibition was blocked with the A2AR antagonists or protein kinase A inhibitor. We also determined that the development of EAMG was accompanied by a T-helper cell imbalance that could be restored following A2AR stimulation that resulted in increased Treg cell levels and a reduction in Th1-, Th2-, and Th17-cell subtypes. An EAMG-preventive treatment regimen was established that consisted of (2-(p-(2-carbonylethyl)phenylethylamino)-5-N-ethylcarboxamidoadenosine) (CGS21680; A2AR agonist) administration 1 day prior to EAMG induction. Administration of CGS21680 29 days post EAMG induction (therapeutic treatment) also ameliorated disease severity. We conclude that A2AR agonists may represent a new class of compounds that can be developed for use in the treatment of myasthenia gravis or other T-cell- and B-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Na Li
- Department of Neurobiology, Harbin Medical University Provincial Key Lab of Neurobiology, Harbin Medical University, Heilongjiang, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|