1
|
Li X, Zhao C. Interleukin-6 in neuroimmunological disorders: Pathophysiology and therapeutic advances with satralizumab. Autoimmun Rev 2025; 24:103826. [PMID: 40324548 DOI: 10.1016/j.autrev.2025.103826] [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: 02/04/2025] [Revised: 05/01/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Interleukin-6 (IL-6) is a multifunctional cytokine produced by various cells of the innate and adaptive immune systems. It acts as a regulatory factor in immunity, inflammation, metabolism, and cellular function in multiple organs and systems. The functionality of IL-6 is achieved through multiple signal transduction pathways, such as the JAK/STAT and the NF-κB signaling pathways. In this review, we highlighted the inflammatory and non-inflammatory functions of IL-6, as well as the associated signaling pathways. The involvement of IL-6 in neuroimmunological disorders suggests that the interleukin-6 receptor (IL-6R) monoclonal antibody, satralizumab, is a potential therapeutic strategy. Phase III clinical trials have already validated the safety and efficiency of satralizumab in treating neuromyelitis optica spectrum disorders (NMOSD) and acetylcholine receptor (AChR) seropositive generalized myasthenia gravis (gMG). This review aims to elucidate the pathophysiological role of IL-6, and explore the clinical implications of satralizumab in neuroimmunological diseases, providing insights into its potential therapeutic applications.
Collapse
Affiliation(s)
- Xicheng Li
- Department of Neurology and Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders (NCND), China
| | - Chongbo Zhao
- Department of Neurology and Rare Disease Center, Huashan Hospital, Fudan University, Shanghai, China; National Center for Neurological Disorders (NCND), China.
| |
Collapse
|
2
|
Habib AA, Zhao C, Aban I, França MC, José JG, Zu Hörste GM, Klimiec-Moskal E, Pulley MT, Tavolini D, Krumova P, Lennon-Chrimes S, Smith J, Thanei GA, Blondeau K, Vodopivec I, Wolfe GI, Murai H. Safety and efficacy of satralizumab in patients with generalised myasthenia gravis (LUMINESCE): a randomised, double-blind, multicentre, placebo-controlled phase 3 trial. Lancet Neurol 2025; 24:117-127. [PMID: 39862880 DOI: 10.1016/s1474-4422(24)00514-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/18/2024] [Accepted: 12/10/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Evidence from preclinical studies suggests that IL-6 signalling has the potential to modulate immunopathogenic mechanisms upstream of autoantibody effector mechanisms in patients with generalised myasthenia gravis. We aimed to assess the safety and efficacy of satralizumab, a humanised monoclonal antibody targeting the IL-6 receptor, in patients with generalised myasthenia gravis. METHODS LUMINESCE was a randomised, double-blind, placebo-controlled, multicentre, phase 3 study at 105 sites, including hospitals and clinics, globally. Eligible patients were aged 12 years and older, with seropositive generalised myasthenia gravis (autoantibodies to the acetylcholine receptor [AChR-IgG], muscle-specific kinase [MuSK-IgG], or low-density lipoprotein receptor-related protein 4 [LRP4-IgG]), a Myasthenia Gravis Foundation of America severity class II-IV, a Myasthenia Gravis Activities of Daily Living (MG-ADL) score of 5 or more (non-ocular contribution >50%), and use of stable background therapy. Patients were randomly assigned (1:1) with a permuted-block randomisation method to receive subcutaneous satralizumab (120 mg for bodyweight ≤100 kg; 180 mg for bodyweight >100 kg) or placebo at weeks 0, 2, 4, and every 4 weeks thereafter until week 24. Randomisation was stratified according to background therapy, autoantibody type, and geographical region. The primary efficacy endpoint was mean change from baseline in total MG-ADL score at week 24 in the modified intention-to-treat population (all randomised AChR-IgG-positive patients who completed at least one post-baseline MG-ADL assessment). Safety was assessed in all randomly assigned patients who received at least one dose of study drug. The open-label extension was terminated early because of the sponsor's decision to halt further development of satralizumab for treatment of generalised myasthenia gravis. This trial is registered with ClinicalTrials.gov, NCT04963270, and EudraCT, 2020-004436-21. FINDINGS Between Oct 19, 2021, and Aug 15, 2023, 188 patients were randomly assigned to satralizumab (n=96) or placebo (n=92). 166 AChR-IgG-positive patients (80 in the placebo group and 86 in the satralizumab group) were included in the modified intention-to-treat population. At week 24, statistically significant yet small improvements in MG-ADL score were observed with satralizumab versus placebo (adjusted mean -3·59, 95% CI -4·15 to -3·02 vs -2·57, -3·25 to -1·88; difference -1·02, -1·88 to -0·16; p=0·0120). The proportion of patients with at least one adverse event during the double-blind period was slightly higher in patients treated with satralizumab compared with patients treated with placebo (86 [90%] patients vs 67 [73%] patients). Three serious adverse events (in three [3%] patients) were reported in the satralizumab group (pneumonia, pyelonephritis, and increased lipase) compared with nine (in six [7%] patients) serious adverse events in the placebo group (COVID-19, COVID-19 pneumonia, bacterial urinary tract infection, chest pain, back pain, and rosacea). There were no deaths or adverse events of special interest. INTERPRETATION Satralizumab was well tolerated and resulted in small improvements in patient-reported and clinician-reported outcomes compared with placebo at week 24 in patients with AChR-IgG-positive generalised myasthenia gravis. Further research analysing the immunological underpinnings of the observed clinical response to IL-6 signalling inhibition in patients with generalised myasthenia gravis and exploring the role of IL-6 in autoantibody-mediated diseases is warranted. FUNDING F Hoffmann La Roche.
Collapse
Affiliation(s)
- Ali A Habib
- Department of Neurology, University of California, Irvine, CA, USA.
| | - Chongbo Zhao
- Department of Neurology and Rare Disease Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Inmaculada Aban
- Department of Biostatistics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Jorge Gustavo José
- Unit of Demyelinating Diseases, CIMT Tucuman Medical Research Center, Hospital Ángel C Padilla, Tucumán, Argentina
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | | | - Michael T Pulley
- Department of Neurology, University of Florida College of Medicine, Jacksonville, FL, USA
| | - Darío Tavolini
- Unit of Demyelinating Diseases, INECO Neurociencias Oroño, Rosario, Argentina
| | | | | | | | | | - Kathleen Blondeau
- F Hoffmann-La Roche, Basel, Switzerland; Parexel Belgium, Wavre, Belgium
| | | | - Gil I Wolfe
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo/SUNY, Buffalo, NY, USA
| | - Hiroyuki Murai
- Department of Neurology, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
3
|
Cavalcante P, Mantegazza R, Antozzi C. Targeting autoimmune mechanisms by precision medicine in Myasthenia Gravis. Front Immunol 2024; 15:1404191. [PMID: 38903526 PMCID: PMC11187261 DOI: 10.3389/fimmu.2024.1404191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Myasthenia Gravis (MG) is a chronic disabling autoimmune disease caused by autoantibodies to the neuromuscular junction (NMJ), characterized clinically by fluctuating weakness and early fatigability of ocular, skeletal and bulbar muscles. Despite being commonly considered a prototypic autoimmune disorder, MG is a complex and heterogeneous condition, presenting with variable clinical phenotypes, likely due to distinct pathophysiological settings related with different immunoreactivities, symptoms' distribution, disease severity, age at onset, thymic histopathology and response to therapies. Current treatment of MG based on international consensus guidelines allows to effectively control symptoms, but most patients do not reach complete stable remission and require life-long immunosuppressive (IS) therapies. Moreover, a proportion of them is refractory to conventional IS treatment, highlighting the need for more specific and tailored strategies. Precision medicine is a new frontier of medicine that promises to greatly increase therapeutic success in several diseases, including autoimmune conditions. In MG, B cell activation, antibody recycling and NMJ damage by the complement system are crucial mechanisms, and their targeting by innovative biological drugs has been proven to be effective and safe in clinical trials. The switch from conventional IS to novel precision medicine approaches based on these drugs could prospectively and significantly improve MG care. In this review, we provide an overview of key immunopathogenetic processes underlying MG, and discuss on emerging biological drugs targeting them. We also discuss on future direction of research to address the need for patients' stratification in endotypes according with genetic and molecular biomarkers for successful clinical decision making within precision medicine workflow.
Collapse
Affiliation(s)
- Paola Cavalcante
- Neurology 4 – Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Renato Mantegazza
- Neurology 4 – Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Carlo Antozzi
- Neurology 4 – Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Immunotherapy and Apheresis Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
4
|
Miyake S, Serizawa K, Onishi S, Katsura Y, Baba M, Kurasawa M, Tomizawa-Shinohara H, Yorozu K, Matsumoto Y, Noguchi-Sasaki M. IL-6 receptor antibody treatment improves muscle weakness in experimental autoimmune myasthenia gravis mouse model. Front Neurol 2024; 15:1356300. [PMID: 38751878 PMCID: PMC11094227 DOI: 10.3389/fneur.2024.1356300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/16/2024] [Indexed: 05/18/2024] Open
Abstract
Myasthenia gravis (MG) is a chronic autoimmune disease characterized by muscle weakness and fatigue. It is caused by pathological autoantibodies against components expressed at neuromuscular junctions, such as acetylcholine receptor (AChR). Interleukin-6 (IL-6) has been suggested to play a role in the pathogenesis of MG, and IL-6 receptor (IL-6R) antibody treatment may provide a novel therapeutic option. In this study, we investigated the effects of IL-6R antibody treatment in an experimental autoimmune MG (EAMG) mouse model. We demonstrated that IL-6R antibody treatment improved muscle weakness, reduced IgG deposition at neuromuscular junctions, and the levels of AChR autoantibodies in serum. In addition, follicular helper T cells and Th17, plasma cells in lymph nodes were lower in IL-6R antibody treated mice. Our findings suggest that IL-6R blockade may be a novel and effective therapeutic strategy for the treatment of MG.
Collapse
Affiliation(s)
- Shota Miyake
- Product Research Department, Chugai Pharmaceutical Co., Ltd., Yokohama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Jia D, Zhang F, Li H, Shen Y, Jin Z, Shi FD, Zhang C. Responsiveness to Tocilizumab in Anti-Acetylcholine Receptor-Positive Generalized Myasthenia Gravis. Aging Dis 2024; 15:824-830. [PMID: 37450932 PMCID: PMC10917550 DOI: 10.14336/ad.2023.0528] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
Tocilizumab, a humanized IL-6R monoclonal antibody, has been used in autoimmune diseases closely related to humoral immunity. This report aims to evaluate the efficacy and safety in patients with anti-acetylcholine receptor-positive (AChR+) generalized myasthenia gravis (gMG). We performed a prospective, open-label, single-arm study in patients with gMG in a 48-week follow-up. All patients were AChR+ and were given tocilizumab by intravenous infusion at a dose of 8 mg/kg at intervals of 4 weeks. The primary endpoint was mean change from baseline in quantitative MG (QMG) score at week 12. The secondary endpoints were mean changes from baseline in MG activities of daily living (MG-ADL) score, AChR-ab titers, and the dosage of oral prednisone at week 12. At week 48, QMG, MG-ADL, and the use of prednisone were also evaluated. Fourteen gMG patients were enrolled and all of them completed the study. Tocilizumab treatment started 8 (4-192) months after the onset of gMG. During tocilizumab treatment, the QMG score was significantly decreased from 15.5 (interqualile range, 9-26) at baseline to 4 (0-9) at week 12 (p < 0.001). The change of ADL was decreased from 14.5(11-19) at baseline to 4 (0-19) at week 12 (p < 0.001) and the change of AChR-ab titers from 15 (7.5-19) at baseline to 6.8 (11.6-4.3) at week 12 (p < 0.001). The dosage of prednisone decreased from baseline 60 (20-65) mg/d to 30 (30-50) mg/d at week 12 (p < 0.001). By the end of the study, the QMG score was 2 (0-7) and MG-ADL score was 1.5 (0-6). 12 (85.7%) patients achieved minimal manifestations. 4 (28.6%) patients were able to discontinue prednisone. No patients experienced exacerbation at the end of the study. No serious adverse events were observed during follow-up. Tocilizumab treatment was associated with a good clinical response and safety over a 48-week observation period, as evidenced by significant improvements in QMG and MG-ADL.
Collapse
Affiliation(s)
- Dongmei Jia
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| | - Fenghe Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| | - Huining Li
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| | - Yi Shen
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| | - Zhao Jin
- Department of Neurology, Third Central Hospital of Tianjin, Tianjin, China.
| | - Fu-Dong Shi
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| | - Chao Zhang
- Department of Neurology and Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin Medical University Tianjin, China.
| |
Collapse
|
6
|
Abstract
Myasthenia gravis (MG) is a neuromuscular autoimmune disorder characterized by chronic but intermittent fatigue of the eye- and general body muscles. Muscle weakness is caused primarily by the binding of an autoantibody to the acetylcholine receptors, resulting in blockage of normal neuromuscular signal transmission. Studies revealed substantial contributions of different proinflammatory or inflammatory mediators in the pathogenesis of MG. Despite these findings, compared to therapeutic approaches that target autoantibody and complements, only a few therapeutics against key inflammatory molecules have been designed or tested in MG clinical trials. Recent research focuses largely on identifying unknown molecular pathways and novel targets involved in inflammation associated with MG. A well-designed combination or adjunct treatment utilizing one or more selective and validated promising biomarkers of inflammation as a component of targeted therapy may yield better treatment outcomes. This review briefly discusses some preclinical and clinical findings of inflammation associated with MG and current therapy approaches and suggest the potential of targeting important inflammatory marker(s) along with current monoclonal antibody or antibody fragment based targeted therapies directed to a variety of cell surface receptors.
Collapse
|
7
|
Blocking interleukin-23 ameliorates neuromuscular and thymic defects in myasthenia gravis. J Neuroinflammation 2023; 20:9. [PMID: 36639663 PMCID: PMC9837970 DOI: 10.1186/s12974-023-02691-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
Acetylcholine receptor (AChR) myasthenia gravis (MG) is a chronic autoimmune disease characterized by muscle weakness. The AChR+ autoantibodies are produced by B-cells located in thymic ectopic germinal centers (eGC). No therapeutic approach is curative. The inflammatory IL-23/Th17 pathway is activated in the thymus as well as in the blood and the muscle, contributing to the MG pathogenic events. We aimed to study a potential new therapeutic approach that targets IL-23p19 (IL-23) in the two complementary preclinical MG models: the classical experimental MG mouse model (EAMG) based on active immunization and the humanized mouse model featuring human MG thymuses engrafted in NSG mice (NSG-MG). In both preclinical models, the anti-IL-23 treatment ameliorated MG clinical symptoms. In the EAMG, the treatment reduced IL-17 related inflammation, anti-AChR IgG2b antibody production, activated transduction pathway involved in muscle regeneration and ameliorated the signal transduction at the neuromuscular junction. In the NSG-MG model, the treatment reduced pathogenic Th17 cell population and expression of genes involved in eGC stabilization and B-cell development in human MG thymus biopsies. Altogether, these data suggest that a therapy targeting IL-23p19 may promote significant clinical ameliorations in AChR+ MG disease due to concomitant beneficial effects on the thymus and skeletal muscle defects.
Collapse
|
8
|
Vanoli F, Mantegazza R. Antibody Therapies in Autoimmune Neuromuscular Junction Disorders: Approach to Myasthenic Crisis and Chronic Management. Neurotherapeutics 2022; 19:897-910. [PMID: 35165857 PMCID: PMC9294078 DOI: 10.1007/s13311-022-01181-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2022] [Indexed: 02/06/2023] Open
Abstract
Myasthenia gravis (MG) is a neurological autoimmune disorder characterized by muscle weakness and fatigue. It is a B cell-mediated disease caused by pathogenic antibodies directed against various components of the neuromuscular junction (NMJ). Despite the wide range of adverse effects, current treatment is still based on non-specific immunosuppression, particularly on long-term steroid usage. The increasing knowledge regarding the pathogenic mechanisms of MG has however allowed to create more target-specific therapies. A very attractive therapeutic approach is currently offered by monoclonal antibodies (mAbs), given their ability to specifically and effectively target different immunopathological pathways, such as the complement cascade, B cell-related cluster of differentiation (CD) proteins, and the human neonatal Fc receptor (FcRn). Up to now, eculizumab, a C5-directed mAb, has been approved for the treatment of generalized MG (gMG) and efgartigimod, a FcRn inhibitor, has just been approved by the U.S. Food and Drug Administration for the treatment of anti-acetylcholine receptor (AChR) antibody positive gMG. Other mAbs are currently under investigation with encouraging preliminary results, further enriching the new range of therapeutic possibilities for MG. This review article provides an overview of the present status of mAb-based therapies for MG, which offer an exciting promise for better outcomes by setting the basis of a precision medicine approach.
Collapse
Affiliation(s)
- Fiammetta Vanoli
- Neuroimmunology and Neuromuscular Disease Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Human Neurosciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Disease Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| |
Collapse
|
9
|
Bahauddin A, Ivannikov M, Wang Z, Jamaluddin M, Curtis K, Ibtehaj N, Yeager L, Soong L, Fang X, Huda R. Histone Deacetylase Isoforms Differentially Modulate Inflammatory and Autoantibody Responses in a Mouse Model of Myasthenia Gravis. Front Neurol 2022; 12:804113. [PMID: 35222229 PMCID: PMC8866970 DOI: 10.3389/fneur.2021.804113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease characterized by chronic muscle fatigue and weakness caused by autoantibodies and complement-mediated damage at neuromuscular junctions. Histone deacetylases (HDACs) are crucial epigenetic regulators of proinflammatory gene expression; however, it is unclear whether HDACs modulate chronic inflammation or autoantibody production associated with MG pathogenesis. We examined expression profiles and serum levels of key inflammatory cytokines (IL-6 and IL-21) and acetylcholine receptor (AChR)-specific autoantibodies following pharmacological inhibition of key HDAC isoforms in a mouse model of MG. We found that HDAC inhibition significantly reduced the production of IL-6, but not IL-21, in AChR-stimulated PBMCs and splenocytes (n = 5 per group). Trichostatin (pan-HDAC inhibitor) treatment of MG-PBMCs (n = 2) also exhibited reduced production of induced IL-6. Although HDAC1 inhibition lowered IL-6 levels the most, HDAC2 inhibition depleted intracellular IL-6 and markedly reduced serum anti-AChR IgG2b in EAMG mice. The transcriptomic profiling and pathway mapping also revealed that autoimmunity-linked, major cell signaling pathways were differentially altered by HDAC1/2 inhibition. HDAC inhibition-mediated reduction in IL-6 and autoantibody levels also correlated with milder disease and preservation of muscle AChR in the treated mice. Overall, our findings revealed isoform-specific functional variance of HDACs in reducing inflammation and identified HDAC-regulated many genes underlying specific inflammatory and autoantibody pathways in EAMG. Thus, the study provides a rationale for further research to evaluate the HDACs or their gene targets as a potential adjunct treatment for MG.
Collapse
Affiliation(s)
- Afrin Bahauddin
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Maxim Ivannikov
- Optical Microscopy Core, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Zhongying Wang
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mohammad Jamaluddin
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Kyra Curtis
- School of Medicine, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Naazneen Ibtehaj
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Linsey Yeager
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ruksana Huda
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
- *Correspondence: Ruksana Huda
| |
Collapse
|
10
|
Howlett-Prieto Q, Langer C, Rezania K, Soliven B. Modulation of immune responses by bile acid receptor agonists in myasthenia gravis. J Neuroimmunol 2020; 349:577397. [PMID: 32979707 DOI: 10.1016/j.jneuroim.2020.577397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 01/13/2023]
Abstract
Bile acids bind to multiple receptors, including Takeda G protein-coupled receptor 5 (TGR5) and farnesoid-X-receptors alpha (FXRα). We compared the response of PBMCs to the activation of these receptors in healthy controls and myasthenic patients. We found that TGR5 is a more potent negative regulator of T cell cytokine response than FXRα in both groups. In contrast, TGR5 and FXRα agonists elicit distinct B cell responses in myasthenia compared to controls, specifically on the frequency of IL-6+ B cells and regulatory B cells, as well as IL-10 secretion from PBMCs. We propose that TGR5 is a potential therapeutic target in myasthenia.
Collapse
Affiliation(s)
- Quentin Howlett-Prieto
- Department of Neurology, The University of Chicago, Chicago, IL 60637, United States of America
| | - Collin Langer
- Department of Neurology, The University of Chicago, Chicago, IL 60637, United States of America
| | - Kourosh Rezania
- Department of Neurology, The University of Chicago, Chicago, IL 60637, United States of America
| | - Betty Soliven
- Department of Neurology, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
11
|
Zhang QX, Li Y, Jiang SM, Zhang LJ, Yi M, Wang J, Qi Y, Yang L, Yang CS. Increased serum IL-36γ levels are associated with disease severity in myasthenia gravis patients. BMC Neurol 2020; 20:307. [PMID: 32814555 PMCID: PMC7436949 DOI: 10.1186/s12883-020-01885-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 08/10/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Interleukin 36 (IL-36), as a gradually recognized cytokine, is involved in the occurrence and evolution of autoimmune diseases. Nevertheless, the relationship between myasthenia gravis (MG) and IL-36 is rarely reported. METHODS We evaluated the serum levels of IL-36 (IL-36α, IL-36β and IL-36γ) by enzyme-linked immunosorbent assay (ELISA). Further, clinical parameters in 97 MG patients and 49 healthy controls (HCs) were carefully measured. RESULTS Serum IL-36γ levels were significantly elevated in the MG patients compared with the HCs (p < 0.0001). Compared to those in remission, patients in the acute phase exhibited higher levels of IL-36α and IL-36γ (p = 0.038 and p = 0.011, respectively). Furthermore, patients with generalized MG (GMG) exhibited markedly higher serum IL-36γ levels than those with ocular MG (OMG) (p = 0.003). CONCLUSIONS The serum levels of IL-36γ in patients with MG were increased and positively correlated with disease severity and may thus have potential as a serological MG marker.
Collapse
Affiliation(s)
- Qiu-Xia Zhang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yue Li
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shu-Min Jiang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Lin-Jie Zhang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Ming Yi
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jing Wang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yuan Qi
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Li Yang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chun-Sheng Yang
- Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
12
|
Angelopoulou E, Paudel YN, Piperi C. Unraveling the Role of Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Myasthenia Gravis. ACS Chem Neurosci 2020; 11:663-673. [PMID: 32017530 DOI: 10.1021/acschemneuro.9b00678] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune T cell-dependent B cell-mediated disorder of the neuromuscular junction (NMJ) characterized by fluctuating skeletal muscle weakness, most commonly attributed to pathogenic autoantibodies against postsynaptic nicotinic acetylcholine receptors (AChRs). Although MG pathogenesis is well-documented, there are no objective biomarkers that could effectively correlate with disease severity or MG clinical subtypes, and current treatment approaches are often ineffective. The receptor for advanced glycation end products (RAGE) is a multiligand cell-bound receptor highly implicated in proinflammatory responses and autoimmunity. Preclinical evidence demonstrates that RAGE and its ligand S100B are upregulated in rat models of experimental autoimmune myasthenia gravis (EAMG). S100B-mediated RAGE activation has been shown to exacerbate EAMG, by enhancing T cell proinflammatory responses, aggravating T helper (Th) subset imbalance, increasing AChR-specific T cell proliferative capacity, and promoting the production of antibodies against AChRs from the spleen. Soluble sRAGE and esRAGE, acting as decoys of RAGE ligands, are found to be significantly reduced in MG patients. Moreover, MG has been associated with increased serum levels of S100A12, S100B and HMGB1. Several studies have shown that the presence of thymic abnormalities, the onset age of MG, and the duration of the disease may affect the levels of these proteins in MG patients. Herein, we discuss the emerging role of RAGE and its ligands in MG immunopathogenesis, their clinical significance as promising biomarkers, as well as the potential therapeutic implications of targeting RAGE signaling in MG treatment.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 46150 Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
13
|
Huda R. New Approaches to Targeting B Cells for Myasthenia Gravis Therapy. Front Immunol 2020; 11:240. [PMID: 32153573 PMCID: PMC7047318 DOI: 10.3389/fimmu.2020.00240] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/29/2020] [Indexed: 01/06/2023] Open
Abstract
Current therapies for myasthenia gravis (MG) are limited, and many investigations have recently focused on target-specific therapies. B cell-targeting monoclonal antibody (mAb) therapies for MG are increasingly attractive due to their specificity and efficacy. The targeted B cell biomarkers are mainly the cluster of differentiation (CD) proteins that mediate maturation, differentiation, or survival of pathogenic B cells. Additional B cell-directed therapies include non-specific peptide inhibitors that preferentially target specific B cell subsets. The primary goals of such therapies are to intercept autoantibodies and prevent the generation of an inflammatory response that contributes to the pathogenesis of MG. Treatment of patients with MG using B cell-directed mAbs, antibody fragments, or selective inhibitors have exhibited moderate to high efficacy in early studies, and some of these therapies appear to be highly promising for further drug development. Numerous other biologics targeting various B cell surface molecules have been approved for the treatment of other conditions or are either in clinical trials or preclinical development stages. These approaches remain to be tested in patients with MG or animal models of the disease. This review article provides an overview of B cell-targeted treatments for MG, including those already available and those still in preclinical and clinical development. We also discuss the potential benefits as well as the shortcomings of these approaches to development of new therapies for MG and future directions in the field.
Collapse
Affiliation(s)
- Ruksana Huda
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
14
|
Beecher G, Putko BN, Wagner AN, Siddiqi ZA. Therapies Directed Against B-Cells and Downstream Effectors in Generalized Autoimmune Myasthenia Gravis: Current Status. Drugs 2019; 79:353-364. [PMID: 30762205 DOI: 10.1007/s40265-019-1065-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Myasthenia gravis is a rare, heterogeneous, classical autoimmune disease characterized by fatigable skeletal muscle weakness, which is directly mediated by autoantibodies targeting various components of the neuromuscular junction, including the acetylcholine receptor, muscle specific tyrosine kinase, and lipoprotein-related protein 4. Subgrouping of myasthenia gravis is dependent on the age of onset, pattern of clinical weakness, autoantibody detected, type of thymic pathology, and response to immunotherapy. Generalized immunosuppressive therapies are effective in all subgroups of myasthenia gravis; however, approximately 15% remain refractory and more effective treatments with improved safety profiles are needed. In recent years, successful utilization of targeted B-cell therapies in this disease has triggered renewed focus in unraveling the underlying immunopathology in attempts to identify newer therapeutic targets. While myasthenia gravis is predominantly B-cell mediated, T cells, T cell-B cell interactions, and B-cell-related factors are increasingly recognized to play key roles in its immunopathology, particularly in autoantibody production, and novel therapies have focused on targeting these specific immune system components. This overview describes the current understanding of myasthenia gravis immunopathology before discussing B-cell-related therapies, their therapeutic targets, and the rationale and evidence for their use. Several prospective studies demonstrated efficacy of rituximab in various myasthenia gravis subtypes, particularly that characterized by antibodies against muscle-specific tyrosine kinase. However, a recent randomized control trial in patients with acetylcholine receptor antibodies was negative. Eculizumab, a complement inhibitor, has recently gained regulatory approval for myasthenia gravis based on a phase III trial that narrowly missed its primary endpoint while achieving robust results in all secondary endpoints. Zilucoplan is a subcutaneously administered terminal complement inhibitor that recently demonstrated significant improvements in functional outcome measures in a phase II trial. Rozanolixizumab, CFZ533, belimumab, and bortezomib are B-cell-related therapies that are in the early stages of evaluation in treating myasthenia gravis. The rarity of myasthenia gravis, heterogeneity in its clinical manifestations, and variability in immunosuppressive regimens are challenges to conducting successful trials. Nonetheless, these are promising times for myasthenia gravis, as renewed research efforts provide novel insights into its immunopathology, allowing for development of targeted therapies with increased efficacy and safety.
Collapse
Affiliation(s)
- Grayson Beecher
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta Hospital, 7-112 Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Brendan Nicholas Putko
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta Hospital, 7-112 Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Amanda Nicole Wagner
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta Hospital, 7-112 Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada
| | - Zaeem Azfer Siddiqi
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta Hospital, 7-112 Clinical Sciences Building, 11350-83 Ave, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
15
|
Cai X, Li Z, Xi J, Song H, Liu J, Zhu W, Guo Y, Jiao Z. Myasthenia gravis and specific immunotherapy: monoclonal antibodies. Ann N Y Acad Sci 2019; 1452:18-33. [PMID: 31393614 DOI: 10.1111/nyas.14195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/23/2019] [Accepted: 06/25/2019] [Indexed: 01/21/2023]
Affiliation(s)
- Xiao‐Jun Cai
- Department of Clinical Pharmacy, Huashan HospitalFudan University Shanghai P. R. China
- Department of Pharmacythe Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi P. R. China
| | - Zai‐Wang Li
- Department of Neurology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan Universitythe First Affiliated Hospital of Southern University of Science and Technology Shenzhen P. R. China
- Department of Neurologythe Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi P. R. China
| | - Jian‐Ying Xi
- Department of Neurology, Huashan HospitalFudan University Shanghai P. R. China
| | - Hui‐Zhu Song
- Department of Pharmacythe Affiliated Wuxi People's Hospital of Nanjing Medical University Wuxi P. R. China
| | - Jue Liu
- Department of Clinical Pharmacy, Huashan HospitalFudan University Shanghai P. R. China
| | - Wen‐Hua Zhu
- Department of Neurology, Huashan HospitalFudan University Shanghai P. R. China
| | - Yi Guo
- Department of Neurology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan Universitythe First Affiliated Hospital of Southern University of Science and Technology Shenzhen P. R. China
| | - Zheng Jiao
- Department of Clinical Pharmacy, Huashan HospitalFudan University Shanghai P. R. China
| |
Collapse
|
16
|
Abstract
Acquired Myasthenia Gravis (MG) is a neuromuscular disease caused by autoantibodies against components of the neuromuscular junction. It is a prototype organ-specific autoimmune disease with well-defined antigenic targets mainly the nicotinic acetylcholine receptor (AChR). Patients suffer from fluctuating, fatigable muscle weakness that worsens with activity and improves with rest. Various therapeutic strategies have been used over the years to alleviate MG symptoms. These strategies aim at improving the transmission of the nerve impulse to muscle or at lowering the immune system with steroids or immunosuppressant drugs. Nevertheless, MG remains a chronic disease and symptoms tend to persist in many patients, some being or becoming refractory over time. In this review, based on recent experimental data on MG or based on results from clinical trials for other autoimmune diseases, we explore new potential therapeutic approaches for MG patients, going from non-specific approaches with the use of stem cells with their anti-inflammatory and immunosuppressive properties to targeted therapies using monoclonal antibodies specific for cell-surface antigens or circulating molecules.
Collapse
Affiliation(s)
- Anthony Behin
- APHP, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France, Institut de Myologie, GH Pitié-Salpêtrière, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Rozen Le Panse
- INSERM U974, Paris, France.,UPMC Sorbonne Université, Paris, France.,AIM, Institut de Myologie, Paris, France
| |
Collapse
|
17
|
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
|
18
|
Jones GW, Hill DG, Cardus A, Jones SA. IL-27: a double agent in the IL-6 family. Clin Exp Immunol 2018; 193:37-46. [PMID: 29437229 DOI: 10.1111/cei.13116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2018] [Indexed: 12/31/2022] Open
Abstract
The cytokine interleukin (IL)-6 is a major therapeutic target for the treatment of various inflammatory and autoimmune diseases. While IL-6 receives considerable attention in studies of innate and adaptive immunity, the IL-6-related family member IL-27 is recognized increasingly for its effects on cellular proliferation, differentiation and leucocyte effector functions. Both cytokines activate responses in myeloid and stromal tissue cells, where they direct the transition from innate to adaptive immunity. However, they are identified frequently as lymphokines that control responses in T cells and B cells. In this regard, IL-27 often opposes the action of IL-6. Here, we will review the role of IL-6 and IL-27 in inflammation, with a particular focus on inflammatory arthritis, and discuss their importance in the diagnosis, stratification and treatment of autoimmune disease.
Collapse
Affiliation(s)
- G W Jones
- Division of Infection and Immunity, The School of Medicine, Systems Immunity University Research Institute, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, UK
| | - D G Hill
- Division of Infection and Immunity, The School of Medicine, Systems Immunity University Research Institute, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, UK
| | - A Cardus
- Division of Infection and Immunity, The School of Medicine, Systems Immunity University Research Institute, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, UK
| | - S A Jones
- Division of Infection and Immunity, The School of Medicine, Systems Immunity University Research Institute, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
19
|
Villegas JA, Van Wassenhove J, Le Panse R, Berrih-Aknin S, Dragin N. An imbalance between regulatory T cells and T helper 17 cells in acetylcholine receptor-positive myasthenia gravis patients. Ann N Y Acad Sci 2018; 1413:154-162. [PMID: 29405352 DOI: 10.1111/nyas.13591] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/15/2022]
Abstract
A chronic autoimmune disease, myasthenia gravis (MG) is characterized in 85% of patients by antibodies directed against the acetylcholine receptor (AChR) located at the neuromuscular junction. The functional and effective balance between regulatory T cells (Treg cells) and effector T cells (Teff cells) is lost in the hyperplastic thymus of MG patients with antibodies specific for the AChR (AChR+ MG patients). The objective of this review is to describe how Treg cells and inflammatory T cells participate in this imbalance and contribute to induce a chronic inflammatory state in the MG thymus. We discuss the origins and characteristics of Treg cells and their reported dysfunctions in AChR+ MG patients. We also review the inflammatory condition observed in MG thymus, including overexpression of interleukin (IL)-1β, IL-6, and IL-23, cytokines that promote the differentiation of T helper 17 (TH 17) cells and the expression of IL-17. We summarize the preclinical models used to determine the implication of expression of cytokines, such as IL-6, IL-12 (IL-23 subunit), IL-17, and interferon γ to the development of experimental autoimmune MG. Finally, we suggest that biological agents, such as humanized monoclonal antibodies that target the IL-23/TH 17 pathway, should be investigated in the context of MG, as they have proven efficiency in other autoimmune diseases.
Collapse
Affiliation(s)
- Jose Adolfo Villegas
- UPMC Sorbonne Universities, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Jérôme Van Wassenhove
- UPMC Sorbonne Universities, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Rozen Le Panse
- UPMC Sorbonne Universities, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Sonia Berrih-Aknin
- UPMC Sorbonne Universities, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France
| | - Nadine Dragin
- UPMC Sorbonne Universities, Paris, France.,INSERM U974, Paris, France.,AIM, Institute of Myology, Paris, France.,Inovarion, Paris, France
| |
Collapse
|
20
|
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
|
21
|
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]
|
22
|
Aguilo-Seara G, Xie Y, Sheehan J, Kusner LL, Kaminski HJ. Ablation of IL-17 expression moderates experimental autoimmune myasthenia gravis disease severity. Cytokine 2017; 96:279-285. [PMID: 28599246 DOI: 10.1016/j.cyto.2017.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/17/2017] [Accepted: 05/08/2017] [Indexed: 01/25/2023]
Abstract
An array of cytokines influences the pathogenesis of early onset myasthenia gravis (MG) and its animal model, experimental autoimmune myasthenia gravis (EAMG). Patients with MG, in particular those with more severe weakness, have elevations of the pro-inflammatory cytokine IL-17 in the blood. We assessed the role of IL-17A in autoimmunity by inducing EAMG in mice with knockout of IL-17 and found a reduction of EAMG severity, but not a complete ablation of disease. The IL-17ko mice had no evidence of weakness, low levels of acetylcholine receptor antibodies, and retention of acetylcholine receptor at the neuromuscular junction. Splenic germinal center size was reduced in EAMG IL-17ko mice along with elevations of Foxp3 and BCL-6 gene expression, suggesting a shift away from pro-inflammatory signals. The results emphasize the importance of IL-17 in EAMG development and that IL-17 independent pathways drive the autoimmune reaction.
Collapse
Affiliation(s)
| | - Yanchen Xie
- Departments of Neurology, George Washington University, Washington, DC, USA
| | - Jarrod Sheehan
- Pharmacology & Physiology, George Washington University, Washington, DC, USA
| | - Linda L Kusner
- Departments of Neurology, George Washington University, Washington, DC, USA; Pharmacology & Physiology, George Washington University, Washington, DC, USA
| | - Henry J Kaminski
- Departments of Neurology, George Washington University, Washington, DC, USA; Pharmacology & Physiology, George Washington University, Washington, DC, USA.
| |
Collapse
|
23
|
Ulusoy C, Çavuş F, Yılmaz V, Tüzün E. Immunization with Recombinantly Expressed LRP4 Induces Experimental Autoimmune Myasthenia Gravis in C57BL/6 Mice. Immunol Invest 2017; 46:490-499. [PMID: 28375749 DOI: 10.1080/08820139.2017.1299754] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Myasthenia gravis (MG) is an autoimmune disease of the neuromuscular junction (NMJ), characterized with muscle weakness. While MG develops due to acetylcholine receptor (AChR) antibodies in most patients, antibodies to muscle-specific receptor tyrosine kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4) may also be identified. Experimental autoimmune myasthenia gravis (EAMG) has been previously induced by both LRP4 immunization and passive transfer of LRP4 antibodies. OBJECTIVE Our aim was to confirm previous results and to test the pathogenic effects of LRP4 immunization in a commonly used mouse strain C57BL/6 (B6) using a recombinantly expressed human LRP4 protein. METHODS B6 mice were immunized with human LRP4 in CFA, Torpedo Californica AChR in CFA or only CFA. Clinical and pathogenic aspects of EAMG were compared among groups. RESULTS LRP4- and AChR-immunized mice showed comparable EAMG clinical severity. LRP4-immunized mice displayed serum antibodies to LRP4 and NMJ IgG and complement factor C3 deposits. IgG2 was the dominant anti-LRP4 isotype. Cultured lymph node cells of LRP4- and AChR-immunized mice gave identical pro-inflammatory cytokine (IL-6, IFN-γ and IL-17) responses to LRP4 and AChR stimulation, respectively. CONCLUSION Our results confirm the EAMG-inducing action of LRP4 immunization and identify B6 as a LRP4-EAMG-susceptible mouse strain. Demonstration of complement fixing anti-LRP4 antibodies in sera and complement/IgG deposits at the NMJ of LRP4-immunized mice indicates complement activation as a putative pathogenic mechanism. We have thus developed a practical LRP4-induced EAMG model using a non-conformational protein and a widely available mouse strain for future investigation of LRP4-related MG.
Collapse
Affiliation(s)
- Canan Ulusoy
- a Department of Neuroscience , Aziz Sancar Institute for Experimental Medical Research, Istanbul Faculty of Medicine, Istanbul University , Istanbul , Turkey
| | - Filiz Çavuş
- b Department of Genetics, Aziz Sancar Institute for Experimental Medical Research, Istanbul Faculty of Medicine , Istanbul University , Istanbul , Turkey
| | - Vuslat Yılmaz
- a Department of Neuroscience , Aziz Sancar Institute for Experimental Medical Research, Istanbul Faculty of Medicine, Istanbul University , Istanbul , Turkey
| | - Erdem Tüzün
- a Department of Neuroscience , Aziz Sancar Institute for Experimental Medical Research, Istanbul Faculty of Medicine, Istanbul University , Istanbul , Turkey
| |
Collapse
|
24
|
Zhang CJ, Gong Y, Zhu W, Qi Y, Yang CS, Fu Y, Chang G, Li Y, Shi S, Wood K, Ladha S, Shi FD, Liu Q, Yan Y. Augmentation of Circulating Follicular Helper T Cells and Their Impact on Autoreactive B Cells in Myasthenia Gravis. THE JOURNAL OF IMMUNOLOGY 2016; 197:2610-7. [DOI: 10.4049/jimmunol.1500725] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/22/2016] [Indexed: 11/19/2022]
|
25
|
Guptill JT, Soni M, Meriggioli MN. Current Treatment, Emerging Translational Therapies, and New Therapeutic Targets for Autoimmune Myasthenia Gravis. Neurotherapeutics 2016; 13:118-31. [PMID: 26510558 PMCID: PMC4720661 DOI: 10.1007/s13311-015-0398-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease associated with the production of autoantibodies against 1) the skeletal muscle acetylcholine receptor; 2) muscle-specific kinase, a receptor tyrosine kinase critical for the maintenance of neuromuscular synapses; 3) low-density lipoprotein receptor-related protein 4, an important molecular binding partner for muscle-specific kinase; and 4) other muscle endplate proteins. In addition to the profile of autoantibodies, MG may be classified according the location of the affected muscles (ocular vs generalized), the age of symptom onset, and the nature of thymic pathology. Immunopathologic events leading to the production of autoantibodies differ in the various disease subtypes. Advances in our knowledge of the immunopathogenesis of the subtypes of MG will allow for directed utilization of the ever-growing repertoire of therapeutic agents that target distinct nodes in the immune pathway relevant to the initiation and maintenance of autoimmune disease. In this review, we examine the pathogenesis of MG subtypes, current treatment options, and emerging new treatments and therapeutic targets.
Collapse
Affiliation(s)
- Jeffrey T Guptill
- Neuromuscular Division, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Madhu Soni
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Matthew N Meriggioli
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
26
|
Ulusoy C, Zibandeh N, Yıldırım S, Trakas N, Zisimopoulou P, Küçükerden M, Tașlı H, Tzartos S, Göker K, Tüzün E, Akkoç T. Dental follicle mesenchymal stem cell administration ameliorates muscle weakness in MuSK-immunized mice. J Neuroinflammation 2015; 12:231. [PMID: 26646841 PMCID: PMC4673854 DOI: 10.1186/s12974-015-0451-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/04/2015] [Indexed: 01/09/2023] Open
Abstract
Background Myasthenia gravis (MG) is an antibody-mediated autoimmune disease of the neuromuscular junction (NMJ), mostly associated with acetylcholine receptor (AChR) antibodies. Around 5–10 % of MG patients show antibodies to muscle-specific tyrosine kinase (MuSK). Mesenchymal stem cell (MSC) administration has been shown to ameliorate muscle weakness in the experimental autoimmune myasthenia gravis (EAMG) model induced by AChR immunization. Methods To investigate the efficacy of stem cell treatment in MuSK-related EAMG, clinical and immunological features of MuSK-immunized mice with or without dental follicle MSC (DFMSC) treatment were compared. Results MuSK-immunized mice intravenously treated with DFMSC after second and third immunizations showed significantly lower EAMG incidence and severity and reduced serum anti-MuSK antibody, NMJ IgG, and C3 deposit levels and CD11b+ lymph node cell ratios. Moreover, lymph node cells of DFMSC-administered mice showed reduced proliferation and IL-6 and IL-12 production responses to MuSK stimulation. By contrast, proportions of B and T cell populations and production of a wide variety of cytokines were not affected from DFMSC treatment. Conclusions Our results suggest that DFMSC treatment shows its beneficial effects mostly through suppression of innate immune system, whereas other immune functions appear to be preserved. Stem cell treatment might thus constitute a specific and effective treatment method in MuSK-associated MG. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0451-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Canan Ulusoy
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Noushin Zibandeh
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Selin Yıldırım
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Nikolaos Trakas
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | | | - Melike Küçükerden
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Hatice Tașlı
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey
| | - Socrates Tzartos
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Kamil Göker
- Department of Oral and Maxillofacial Surgery, Marmara University Faculty of Dentistry, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Institute for Experimental Medical Research (DETAE), Istanbul University, Istanbul, Turkey. .,Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Capa, Turkey.
| | - Tunç Akkoç
- Division of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
27
|
Guidelines for standard preclinical experiments in the mouse model of myasthenia gravis induced by acetylcholine receptor immunization. Exp Neurol 2015; 270:11-7. [DOI: 10.1016/j.expneurol.2015.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/13/2015] [Accepted: 02/04/2015] [Indexed: 01/25/2023]
|
28
|
Zhang DQ, Wang R, Li T, Li X, Qi Y, Wang J, Yang L. Remarkably increased resistin levels in anti-AChR antibody-positive myasthenia gravis. J Neuroimmunol 2015; 283:7-10. [DOI: 10.1016/j.jneuroim.2015.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 11/30/2022]
|
29
|
Huda R, Strait RT, Tüzün E, Finkelman FD, Christadoss P. IgG1 deficiency exacerbates experimental autoimmune myasthenia gravis in BALB/c mice. J Neuroimmunol 2015; 281:68-72. [PMID: 25867470 PMCID: PMC4395862 DOI: 10.1016/j.jneuroim.2015.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
Abstract
Myasthenia gravis is an autoimmune disease characterized by muscle weakness due to neuromuscular junction (NMJ) damage by anti-acetylcholine receptor (AChR) auto-antibodies and complement. In experimental autoimmune myasthenia gravis (EAMG), which is induced by immunization with Torpedo AChR in CFA, anti-AChR IgG2b and IgG1 are the predominant isotypes in the circulation. Complement activation by isotypes such as IgG2b plays a crucial role in EAMG pathogenesis; this suggested the possibility that IgG1, which does not activate complement through the classical pathway, may suppress EAMG. In this study, we show that AChR-immunized BALB/c mice genetically deficient for IgG1 produce higher levels of complement-activating isotypes of anti-AChR, especially IgG3 and IgG2a, and develop increased IgG3/IgG2a deposits at the NMJ, as compared to wild type (WT) BALB/c mice. Consistent with this, AChR-immunized IgG1(-/-) BALB/c mice lose muscle strength and muscle AChR to a greater extent than AChR-immunized WT mice. These observations demonstrate that IgG1 deficiency leads to increased severity of EAMG associated with an increase in complement activating IgG isotypes. Further studies are needed to dissect the specific role or mechanism of IgG1 in limiting EAMG and that of EAMG exacerbating role of complement activating IgG3 and IgG2a in IgG1 deficiency.
Collapse
Affiliation(s)
- Ruksana Huda
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA.
| | - Richard T Strait
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Erdem Tüzün
- Department of Neuroscience, Institute for Experimental Medical Research, University of Istanbul, Istanbul 34390, Turkey
| | - Fred D Finkelman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA; Department of Medicine, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220, USA
| | - Premkumar Christadoss
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA.
| |
Collapse
|
30
|
Schaffert H, Pelz A, Saxena A, Losen M, Meisel A, Thiel A, Kohler S. IL-17-producing CD4(+) T cells contribute to the loss of B-cell tolerance in experimental autoimmune myasthenia gravis. Eur J Immunol 2015; 45:1339-47. [PMID: 25676041 DOI: 10.1002/eji.201445064] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 01/07/2015] [Accepted: 02/09/2015] [Indexed: 01/01/2023]
Abstract
The role of Th17 cells in the pathogenesis of autoantibody-mediated diseases is unclear. Here, we assessed the contribution of Th17 cells to the pathogenesis of experimental autoimmune myasthenia gravis (EAMG), which is induced by repetitive immunizations with Torpedo californica acetylcholine receptor (tAChR). We show that a significant fraction of tAChR-specific CD4(+) T cells is producing IL-17. IL-17(ko) mice developed fewer or no EAMG symptoms, although the frequencies of tAChR-specific CD4(+) T cells secreting IL-2, IFN-γ, or IL-21, and the percentage of FoxP3(+) Treg cells were similar to WT mice. Even though the total anti-tAChR antibody levels were equal, the complement fixating IgG2b subtype was reduced in IL-17(ko) as compared to WT mice. Most importantly, pathogenic anti-murine AChR antibodies were significantly lower in IL-17(ko) mice. Furthermore, we confirmed the role of Th17 cells in EAMG pathogenesis by the reconstitution of TCR β/δ(ko) mice with WT or IL-17(ko) CD4(+) T cells. In conclusion, we show that the level of IgG2b and the loss of B-cell tolerance, which results in pathogenic anti-murine AChR-specific antibodies, are dependent on IL-17 production by CD4(+) T cells. Thus, we describe here for the first time how Th17 cells are involved in the induction of classical antibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Hanne Schaffert
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany
| | - Andreas Pelz
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany
| | - Abhishek Saxena
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Andreas Meisel
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany.,NeuroCure Clinical Research Center (NCRC), Charité-University Medicine Berlin, Berlin, Germany.,Department of Neurology, Charité-University Medicine Berlin, Berlin, Germany
| | - Andreas Thiel
- Department of Regenerative Immunology and Aging, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Siegfried Kohler
- Department of Experimental Neurology, Charité-University Medicine Berlin, Berlin, Germany.,NeuroCure Clinical Research Center (NCRC), Charité-University Medicine Berlin, Berlin, Germany.,Department of Neurology, Charité-University Medicine Berlin, Berlin, Germany
| |
Collapse
|
31
|
IL-6 and Akt are involved in muscular pathogenesis in myasthenia gravis. Acta Neuropathol Commun 2015; 3:1. [PMID: 25627031 PMCID: PMC4308930 DOI: 10.1186/s40478-014-0179-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 12/15/2014] [Indexed: 01/11/2023] Open
Abstract
Introduction Anti-acetylcholine receptor (AChR) autoantibodies target muscles in spontaneous human myasthenia gravis (MG) and its induced experimental autoimmune model MG (EAMG). The aim of this study was to identify novel functional mechanisms occurring in the muscle pathology of myasthenia. Results A transcriptome analysis performed on muscle tissue from MG patients (compared with healthy controls) and from EAMG rats (compared with control rats) revealed a deregulation of genes associated with the Interleukin-6 (IL-6) and Insulin-Like Growth Factor 1 (IGF-1) pathways in both humans and rats. The expression of IL-6 and its receptor IL-6R transcripts was found to be altered in muscles of EAMG rats and mice compared with control animals. In muscle biopsies from MG patients, IL-6 protein level was higher than in control muscles. Using cultures of human muscle cells, we evaluated the effects of anti-AChR antibodies on IL-6 production and on the phosphorylation of Protein Kinase B (PKB/Akt). Most MG sera and some monoclonal anti-AChR antibodies induced a significant increase in IL-6 production by human muscle cells. Furthermore, Akt phosphorylation in response to insulin was decreased in the presence of monoclonal anti-AChR antibodies. Conclusions Anti-AChR antibodies alter IL-6 production by muscle cells, suggesting a putative novel functional mechanism of action for the anti-AChR antibodies. IL-6 is a myokine with known effects on signaling pathways such as Akt/mTOR (mammalian Target of Rapamycin). Since Akt plays a key role in multiple cellular processes, the reduced phosphorylation of Akt by the anti-AChR antibodies may have a significant impact on the muscle fatigability observed in MG patients. Electronic supplementary material The online version of this article (doi:10.1186/s40478-014-0179-6) contains supplementary material, which is available to authorized users.
Collapse
|
32
|
Ulusoy C, Kim E, Tüzün E, Huda R, Yılmaz V, Poulas K, Trakas N, Skriapa L, Niarchos A, Strait RT, Finkelman FD, Turan S, Zisimopoulou P, Tzartos S, Saruhan-Direskeneli G, Christadoss P. Preferential production of IgG1, IL-4 and IL-10 in MuSK-immunized mice. Clin Immunol 2014; 151:155-63. [DOI: 10.1016/j.clim.2014.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 02/06/2014] [Accepted: 02/20/2014] [Indexed: 01/09/2023]
|
33
|
Aguzzi A, Kranich J, Krautler NJ. Follicular dendritic cells: origin, phenotype, and function in health and disease. Trends Immunol 2013; 35:105-13. [PMID: 24315719 DOI: 10.1016/j.it.2013.11.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 10/31/2013] [Accepted: 11/07/2013] [Indexed: 01/15/2023]
Abstract
Follicular dendritic cells (FDCs) were originally identified by their specific morphology and by their ability to trap immune-complexed antigen in B cell follicles. By virtue of the latter as well as the provision of chemokines, adhesion molecules, and trophic factors, FDCs participate in the shaping of B cell responses. Importantly, FDCs also supply tingible body macrophages (TBMs) with the eat-me-signaling molecule milk fat globule-EGF factor 8 (Mfge8), thereby enabling the disposal of apoptotic B cells. Recent studies have provided fundamental insights into the multiple functions of FDCs in both physiological and pathophysiological contexts and into their origin. Here we review these findings, and discuss current concepts related to FDC histogenesis both in lymphoid organs and in inflammatory lymphoneogenesis.
Collapse
Affiliation(s)
- Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland.
| | - Jan Kranich
- Institute for Immunology, Ludwig Maximilians University, Munich, Germany
| | - Nike Julia Krautler
- Garvan Institute of Medical Research, Darlinghurst, New South Wales, Sydney, Australia.
| |
Collapse
|
34
|
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]
|
35
|
Jakubíková M, Piťha J, Latta J, Ehler E, Schutzner J. Myasthenia gravis, Castleman disease, pemphigus, and anti-phospholipid syndrome. Muscle Nerve 2013; 47:447-51. [PMID: 23386221 DOI: 10.1002/mus.23657] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2012] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Myasthenia gravis is an autoimmune disease marked by neuromuscular transmission failure at the neuromuscular junction. Castleman disease is a rare lymphoproliferative disease characterized by non-cancerous angiofolicular hyperplasia of lymphatic tissue. METHODS AND RESULTS We describe a young man with rapid, successive manifestations of myasthenia gravis, a solitary form of Castleman disease, pemphigus vulgaris, and anti-phospholipid syndrome, which resulted in 2 ischemic cerebrovascular events that caused a severe central neurological deficit. DISCUSSION We were unable to find a similar case in the literature, but we hypothesize that the temporal concidence of these clinical entities may be related to a common immunological pathway, such as B-cell activation. Therefore, we treated the patient with an immunosuppressant and anticoagulant treatment, as well as rituximab, a monoclonal antibody therapy against CD20+.
Collapse
Affiliation(s)
- Michala Jakubíková
- Department of Neurology and Center of Clinical Neuroscience, Charles University Prague, First Faculty of Medicine and General University Hospital, Prague Kateřinská 30, 128 01 Praha 2, Czech Republic.
| | | | | | | | | |
Collapse
|
36
|
Wu B, Goluszko E, Huda R, Tüzün E, Christadoss P. Experimental Autoimmune Myasthenia Gravis in the Mouse. ACTA ACUST UNITED AC 2013; Chapter 15:Unit 15.8.. [DOI: 10.1002/0471142735.im1508s100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Bo Wu
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Elzbieta Goluszko
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Ruksana Huda
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Erdem Tüzün
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Premkumar Christadoss
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| |
Collapse
|
37
|
Samavedam UKS, Kalies K, Scheller J, Sadeghi H, Gupta Y, Jonkman MF, Schmidt E, Westermann J, Zillikens D, Rose-John S, Ludwig RJ. Recombinant IL-6 treatment protects mice from organ specific autoimmune disease by IL-6 classical signalling-dependent IL-1ra induction. J Autoimmun 2013; 40:74-85. [DOI: 10.1016/j.jaut.2012.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 11/17/2022]
|
38
|
Chae CS, Kwon HK, Hwang JS, Kim JE, Im SH. Prophylactic effect of probiotics on the development of experimental autoimmune myasthenia gravis. PLoS One 2012; 7:e52119. [PMID: 23284891 PMCID: PMC3527378 DOI: 10.1371/journal.pone.0052119] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 11/12/2012] [Indexed: 12/03/2022] Open
Abstract
Probiotics are live bacteria that confer health benefits to the host physiology. Although protective role of probiotics have been reported in diverse diseases, no information is available whether probiotics can modulate neuromuscular immune disorders. We have recently demonstrated that IRT5 probiotics, a mixture of 5 probiotics, could suppress diverse experimental disorders in mice model. In this study we further investigated whether IRT5 probiotics could modulate the progression of experimental autoimmune myasthenia gravis (EAMG). Myasthenia gravis (MG) is a T cell dependent antibody mediated autoimmune disorder in which acetylcholine receptor (AChR) at the neuromuscular junction is the major auto-antigen. Oral administration of IRT5 probiotics significantly reduced clinical symptoms of EAMG such as weight loss, body trembling and grip strength. Prophylactic effect of IRT5 probiotics on EMAG is mediated by down-regulation of effector function of AChR-reactive T cells and B cells. Administration of IRT5 probiotics decreased AChR-reactive lymphocyte proliferation, anti-AChR reactive IgG levels and inflammatory cytokine levels such as IFN-γ, TNF-α, IL-6 and IL-17. Down-regulation of inflammatory mediators in AChR-reactive lymphocytes by IRT5 probiotics is mediated by the generation of regulatory dendritic cells (rDCs) that express increased levels of IL-10, TGF-β, arginase 1 and aldh1a2. Furthermore, DCs isolated from IRT5 probiotics-fed group effectively converted CD4+ T cells into CD4+Foxp3+ regulatory T cells compared with control DCs. Our data suggest that IRT5 probiotics could be applicable to modulate antibody mediated autoimmune diseases including myasthenia gravis.
Collapse
Affiliation(s)
- Chang-Suk Chae
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju, Korea
| | - Ho-Keun Kwon
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju, Korea
| | - Ji-Sun Hwang
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju, Korea
| | - Jung-Eun Kim
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju, Korea
| | - Sin-Hyeog Im
- School of Life Sciences and Immune Synapse Research Center, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju, Korea
- * E-mail:
| |
Collapse
|
39
|
El Shikh MEM, Pitzalis C. Follicular dendritic cells in health and disease. Front Immunol 2012; 3:292. [PMID: 23049531 PMCID: PMC3448061 DOI: 10.3389/fimmu.2012.00292] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/31/2012] [Indexed: 12/17/2022] Open
Abstract
Follicular dendritic cells (FDCs) are unique immune cells that contribute to the regulation of humoral immune responses. These cells are located in the B-cell follicles of secondary lymphoid tissues where they trap and retain antigens (Ags) in the form of highly immunogenic immune complexes (ICs) consisting of Ag plus specific antibody (Ab) and/or complement proteins. FDCs multimerize Ags and present them polyvalently to B-cells in periodically arranged arrays that extensively crosslink the B-cell receptors for Ag (BCRs). FDC-FcγRIIB mediates IC periodicity, and FDC-Ag presentation combined with other soluble and membrane bound signals contributed by FDCs, like FDC-BAFF, -IL-6, and -C4bBP, are essential for the induction of the germinal center (GC) reaction, the maintenance of serological memory, and the remarkable ability of FDC-Ags to induce specific Ab responses in the absence of cognate T-cell help. On the other hand, FDCs play a negative role in several disease conditions including chronic inflammatory diseases, autoimmune diseases, HIV/AIDS, prion diseases, and follicular lymphomas. Compared to other accessory immune cells, FDCs have received little attention, and their functions have not been fully elucidated. This review gives an overview of FDC structure, and recapitulates our current knowledge on the immunoregulatory functions of FDCs in health and disease. A better understanding of FDCs should permit better regulation of Ab responses to suit the therapeutic manipulation of regulated and dysregulated immune responses.
Collapse
Affiliation(s)
- Mohey Eldin M El Shikh
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | | |
Collapse
|
40
|
Minogue AM, Barrett JP, Lynch MA. LPS-induced release of IL-6 from glia modulates production of IL-1β in a JAK2-dependent manner. J Neuroinflammation 2012; 9:126. [PMID: 22697788 PMCID: PMC3418561 DOI: 10.1186/1742-2094-9-126] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/14/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Compelling evidence has implicated neuroinflammation in the pathogenesis of a number of neurodegenerative conditions. Chronic activation of both astrocytes and microglia leads to excessive secretion of proinflammatory molecules such as TNF α, IL-6 and IL-1 β with potentially deleterious consequences for neuronal viability. Many signaling pathways involving the mitogen-activated protein kinases (MAPKs), nuclear factor κ B (NF κ B) complex and the Janus kinases (JAKs)/signal transducers and activators of transcription (STAT)-1 have been implicated in the secretion of proinflammatory cytokines from glia. We sought to identify signaling kinases responsible for cytokine production and to delineate the complex interactions which govern time-related responses to lipopolysaccharide (LPS). METHODS We examined the time-related changes in certain signaling events and the release of proinflammatory cytokines from LPS-stimulated co-cultures of astrocytes and microglia isolated from neonatal rats. RESULTS TNF α was detected in the supernatant approximately 1 to 2 hours after LPS treatment while IL-1 β and IL-6 were detected after 2 to 3 and 4 to 6 hours, respectively. Interestingly, activation of NF κ B signaling preceded release of all cytokines while phosphorylation of STAT1 was evident only after 2 hours, indicating that activation of JAK/STAT may be important in the up-regulation of IL-6 production. Additionally, incubation of glia with TNF α induced both phosphorylation of JAK2 and STAT1 and the interaction of JAK2 with the TNF α receptor (TNFR1). Co-treatment of glia with LPS and recombinant IL-6 protein attenuated the LPS-induced release of both TNF α and IL-1 β while potentiating the effect of LPS on suppressor of cytokine signaling (SOCS)3 expression and IL-10 release. CONCLUSIONS These data indicate that TNF α may regulate IL-6 production through activation of JAK/STAT signaling and that the subsequent production of IL-6 may impact on the release of TNF α, IL-1 β and IL-10.
Collapse
Affiliation(s)
- Aedín M Minogue
- Trinity College Institute for Neuroscience, Lloyd building, University of Dublin, Trinity College, College Green, Dublin 2, Ireland.
| | | | | |
Collapse
|
41
|
Croker BA, Kiu H, Pellegrini M, Toe J, Preston S, Metcalf D, O'Donnell JA, Cengia LH, McArthur K, Nicola NA, Alexander WS, Roberts AW. IL-6 promotes acute and chronic inflammatory disease in the absence of SOCS3. Immunol Cell Biol 2012; 90:124-9. [PMID: 21519345 PMCID: PMC3146962 DOI: 10.1038/icb.2011.29] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The lack of expression of the suppressor of cytokine signalling-3 (SOCS3) or inactivation of the negative regulatory capacity of SOCS3 has been well documented in rheumatoid arthritis, viral hepatitis and cancer. The specific qualitative and quantitative consequences of SOCS3 deficiency on interleukin-6 (IL-6)-mediated pro- and anti-inflammatory responses remain controversial in vitro and unknown in vivo. Mice with a conditional deletion of SOCS3 in hematopoietic cells develop lethal inflammatory disease during adult life and develop gross histopathological changes during experimental arthritis, typified by elevated IL-6 levels. To clarify the nature of the IL-6 responses in vivo, we generated mice deficient in SOCS3 (SOCS3(-/Δvav)) or both SOCS3 and IL-6 (IL-6(-/-)/SOCS3(-/Δvav)), and examined responses in models of acute and chronic inflammation. Acute responses to IL-1β were lethal to SOCS3(-/Δvav) mice but not IL-6(-/-)/SOCS3(-/Δvav) mice, indicating that IL-6 was required for the lethal inflammation induced by IL-1β. Administration of IL-1β to SOCS3(-/Δvav) mice induced systemic apoptosis of lymphocytes in the thymus, spleen and lymph nodes that was dependent on the presence of IL-6. IL-6 deficiency prolonged survival of SOCS3(-/Δvav) mice and ameliorated spontaneous inflammatory disease developing during adult life. Infection of SOCS3(-/Δvav) mice with LCMV induced a lethal inflammatory response that was dependent on IL-6, despite SOCS3(-/Δvav) mice controlling viral replication. We conclude that SOCS3 is required for survival during inflammatory responses and is a critical regulator of IL-6 in vivo.
Collapse
Affiliation(s)
- Ben A Croker
- Department of Inflammation, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Baggi F, Antozzi C, Toscani C, Cordiglieri C. Acetylcholine Receptor-Induced Experimental Myasthenia Gravis: What Have We Learned from Animal Models After Three Decades? Arch Immunol Ther Exp (Warsz) 2011; 60:19-30. [DOI: 10.1007/s00005-011-0158-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 09/28/2011] [Indexed: 01/23/2023]
|
43
|
Wu B, Goluszko E, Huda R, Tüzün E, Christadoss P. Experimental Autoimmune Myasthenia Gravis in the Mouse. ACTA ACUST UNITED AC 2011; Chapter 15:Unit 15.23. [PMID: 22048803 DOI: 10.1002/0471142735.im1523s95] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Bo Wu
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Elzbieta Goluszko
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Ruksana Huda
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Erdem Tüzün
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| | - Premkumar Christadoss
- Department of Microbiology and Immunology, University of Texas Medical Branch Galveston Texas
| |
Collapse
|
44
|
Activation of the receptor for advanced glycation end products (RAGE) exacerbates experimental autoimmune myasthenia gravis symptoms. Clin Immunol 2011; 141:36-48. [DOI: 10.1016/j.clim.2011.04.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 04/20/2011] [Accepted: 04/21/2011] [Indexed: 12/20/2022]
|
45
|
Tüzün E, Huda R, Christadoss P. Complement and cytokine based therapeutic strategies in myasthenia gravis. J Autoimmun 2011; 37:136-43. [PMID: 21636248 DOI: 10.1016/j.jaut.2011.05.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 05/02/2011] [Indexed: 01/17/2023]
Abstract
Myasthenia gravis (MG) is a T cell-dependent and antibody-mediated disease in which the target antigen is the skeletal muscle acetylcholine receptor (AChR). In the last few decades, several immunological factors involved in MG pathogenesis have been discovered mostly by studies utilizing the experimental autoimmune myasthenia gravis (EAMG) model. Nevertheless, MG patients are still treated with non-specific global immunosuppression that is associated with severe chronic side effects. Due to the high heterogeneity of AChR epitopes and antibody responses involved in MG pathogenesis, the specific treatment of MG symptoms have to be achieved by inhibiting the complement factors and cytokines involved in anti-AChR immunity. EAMG studies have clearly shown that inhibition of the classical and common complement pathways effectively and specifically diminish the neuromuscular junction destruction induced by anti-AChR antibodies. The inborn or acquired deficiencies of IL-6, TNF-α and TNF receptor functions are associated with the lowest EAMG incidences. Th17-type immunity has recently emerged as an important contributor of EAMG pathogenesis. Overall, these results suggest that inhibition of the complement cascade and the cytokine networks alone or in combination might aid in development of future treatment models that would reduce MG symptoms with highest efficacy and lowest side effect profile.
Collapse
Affiliation(s)
- Erdem Tüzün
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | | | |
Collapse
|
46
|
Na SJ, So SH, Lee KO, Choi YC. Elevated serum level of interleukin-32α in the patients with myasthenia gravis. J Neurol 2011; 258:1865-70. [PMID: 21487807 DOI: 10.1007/s00415-011-6036-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/10/2011] [Accepted: 03/31/2011] [Indexed: 10/18/2022]
Abstract
A new cytokine, interleukin-32 (IL-32), has been implicated in the pro-inflammatory immune responses in several autoimmune disorders, such as rheumatoid arthritis and inflammatory bowel diseases. Myasthenia gravis (MG) is a well-characterized autoimmune disease directed at the postsynaptic acetylcholine receptor (AChR) or end plate of the neuromuscular junction. IL-32 is a cytokine that induces tumor necrosis factor (TNF)-α, IL-6, IL-1β, and chemokine. IL-6, TNF-α, and IL-2 are related to the pathogenesis and immunoregulation of MG. The gene expression of IL-32 is increased in human natural killer (NK) cells and T lymphocytes when stimulated by IL-2 or mitogen. NK cells influence the development of experimental autoimmune MG (EAMG) and possibly MG. The aim of this study was to examine whether IL-32α levels are increased in patients with MG and to investigate the relationship between IL-32α levels and disease activity in human MG. Serum IL-32α levels were significantly higher in the MG patients (p = 0.03): 460.07 ± 192.30 pg/mL in MG patients and 248.45 ± 188.42 pg/mL in the healthy control group. Although there was no significant statistical difference, serum IL-32α levels of patients with both anti-AChR binding and blocking antibodies trended to be higher than those without either antibodies (521.56 ± 212.92 pg/mL vs. 339.52 ± 182.78 pg/mL, p = 0.16). IL-32α serum levels tended to decrease with clinical improvement in generalized MG. This study suggests the possibility that IL-32 might contribute to MG pathogenesis or immunoregulation.
Collapse
Affiliation(s)
- Sang-Jun Na
- Department of Neurology, Gangnam Severance Hospital, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, 146-92 Gangnam-gu, Seoul, 135-720, Republic of Korea
| | | | | | | |
Collapse
|
47
|
Aricha R, Mizrachi K, Fuchs S, Souroujon MC. Blocking of IL-6 suppresses experimental autoimmune myasthenia gravis. J Autoimmun 2011; 36:135-41. [DOI: 10.1016/j.jaut.2010.12.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 11/23/2010] [Accepted: 12/06/2010] [Indexed: 12/17/2022]
|
48
|
Ma LP, Premaratne G, Bollano E, Lindholm C, Fu M. Interleukin-6-deficient mice resist development of experimental autoimmune cardiomyopathy induced by immunization of β1-adrenergic receptor. Int J Cardiol 2011; 155:20-5. [PMID: 21334079 DOI: 10.1016/j.ijcard.2011.01.085] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 01/01/2011] [Indexed: 11/28/2022]
Abstract
BACKGROUND IL-6 is known to be an important mediator in immune response and is now suggested to be involved in the pathogenesis of autoimmune diseases. However, little is known about the role of IL-6 in β(1)-adrenergic receptor induced autoimmune cardiomyopathy. MATERIALS AND METHODS Twenty IL-6-deficient (IL-6(-/-)) mice and fifty-one wild type C57BL/6J (WT) mice were immunized with a synthetic peptide corresponding to the second extracellular loop of the β(1) (β(1)AR ECII) at 0, 1, 5, 9, 13 weeks and observed until 25 weeks. Another forty-one WT mice and twenty IL-6(-/-) mice were used as controls receiving vehicle in the same manner. RESULTS As compared with IL-6(-/-) immunized and control mice, WT immunized mice showed increased end-systolic left ventricular dimension and end-diastolic left ventricular dimension as well as decreased fractional shortening and circumferential fiber shortening in the end of the experiment, which was accompanied by significantly increased antibody level. Moreover, mRNAs encoding for β(1)-adrenergic receptor kinase (GRK2), B-type natriuretic peptide (BNP) and β(1) adrenergic receptor (Adrb1) in heart tissues from WT immunized group were increased. There was a significant positive correlation among end-diastolic left ventricular dimension, autoantibody titer and mRNA expressions of BNP, Adrb1 and GRK2. CONCLUSION Our results demonstrated that immunization with β1AR ECII was unable to induce an early stage phenotype of cardiomyopathy in IL-6(-/-) mice, being different from wild type in which cardiomyopathy was observed, suggesting that IL-6 plays a key role in the regulation of β(1)AR induced autoimmune cardiomyopathy possibly through its enhanced antibody production.
Collapse
Affiliation(s)
- Li-Ping Ma
- Dept of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | | | | | | | | |
Collapse
|
49
|
Kusner LL, Kaminski HJ, Soltys J. Effect of complement and its regulation on myasthenia gravis pathogenesis. Expert Rev Clin Immunol 2010; 4:43-52. [PMID: 20477586 DOI: 10.1586/1744666x.4.1.43] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Myasthenia gravis (MG) is primarily caused by antibodies directed towards the skeletal muscle acetylcholine receptor, leading to muscle weakness. Although these antibodies may induce compromise of neuromuscular transmission by blocking acetylcholine receptor function or antigenic modulation, the predominant mechanism of injury to the neuromuscular junction is complement-mediated lysis of the postsynaptic membrane. The vast majority of data to support the role of complement derives from experimentally acquired MG (EAMG). In this article, we review studies that demonstrate the central role of complement in EAMG and MG pathogenesis along with the emerging role of complement in T- and B-cell function, as well as the potential for complement inhibitor-based therapy to treat human MG.
Collapse
Affiliation(s)
- Linda L Kusner
- Department of Neurology & Psychiatry, Saint Louis University, 1438 South Grand Blvd, St Louis, MO 63104, USA.
| | | | | |
Collapse
|
50
|
Gil M, Park SJ, Chung YS, Park CS. Interleukin-15 enhances proliferation and chemokine secretion of human follicular dendritic cells. Immunology 2010; 130:536-44. [PMID: 20331472 DOI: 10.1111/j.1365-2567.2010.03252.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The germinal centre (GC) is a specialized microenvironment where high-affinity antibodies are produced through hypermutation and isotype switching. Follicular dendritic cells (FDCs) are the stromal cells of the GC. The timely expansion and establishment of an FDC network is essential for a protective GC reaction; however, only a few factors modulating FDC development have been recognized. In this study, we report that interleukin-15 (IL-15) enhances human primary FDC proliferation and regulates cytokine secretion. The FDCs express IL-15 receptor complexes for IL-15 signal transduction as well as for specific binding. Moreover, the secretion of chemokines CCL-2, CCL-5, CXCL-5 and CXCL-8 was reduced by blocking IL-15 signalling while the secretion of other cytokines, and the expression of CD14, CD44, CD54 (ICAM-1) and CD106 (VCAM-1) proteins remained unchanged. These results suggest that IL-15 plays a crucial role in the development of FDC networks during GC reaction, offering a new target for immune modulation.
Collapse
Affiliation(s)
- Minchan Gil
- Department of Pathology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|