1
|
Akatani R, Chihara N, Hara A, Tsuji A, Koto S, Kobayashi K, Toda T, Matsumoto R. Interleukin-6 Signaling Blockade Induces Regulatory Plasmablasts in Neuromyelitis Optica Spectrum Disorder. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200266. [PMID: 38889374 PMCID: PMC11188987 DOI: 10.1212/nxi.0000000000200266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/17/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Interleukin-6 receptor antibodies (IL-6R Abs), including satralizumab, are increasingly used to prevent relapse for neuromyelitis optica spectrum disorder (NMOSD). However, the detailed mechanism of action of this treatment on the lymphocyte phenotype remains unclear. This study focused on B cells in patients with NMOSD, hypothesizing that IL-6R Ab enables B cells to acquire regulatory functions by producing the anti-inflammatory cytokine IL-10. METHODS Peripheral blood mononuclear cells were stimulated in vitro to induce the expansion of B-cell subsets, double-negative B cells (DNs; CD19+ IgD-, CD27-) and plasmablasts (PBs; CD19+, CD27hi, CD38hi). Whole B cells, DNs, or PBs were isolated after culture with IL-6R Ab, and IL-10 expression was quantified using quantitative PCR and a cytometric bead array. RNA sequencing was performed to identify the marker of regulatory PBs induced by IL-6R Ab. RESULTS DNs and PBs were observed to expand in patients with NMSOD during the acute attacks. In the in vitro model, IL-6R Ab increased IL-10 expression in B cells. Notably, IL-10 expression increased in PBs but not in DNs. Using RNA sequencing, CD200 was identified as a marker of regulatory PBs among the differentially expressed upregulated genes. CD200+ PBs produced more IL-10 than CD200- PBs. Furthermore, patients with NMOSD who received satralizumab had a higher proportion of CD200+ PBs than patients during the acute attacks. DISCUSSION Treatment with IL-6 signaling blockade elicited a regulatory phenotype in B cells and PBs. CD200+ PBs may be a marker of treatment responsiveness in the context of NMOSD pathophysiology.
Collapse
Affiliation(s)
- Ritsu Akatani
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Norio Chihara
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Atsushi Hara
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Asato Tsuji
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Shusuke Koto
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Kazuhiro Kobayashi
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Tatsushi Toda
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Riki Matsumoto
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
2
|
Tieck MP, Vasilenko N, Ruschil C, Kowarik MC. Peripheral memory B cells in multiple sclerosis vs. double negative B cells in neuromyelitis optica spectrum disorder: disease driving B cell subsets during CNS inflammation. Front Cell Neurosci 2024; 18:1337339. [PMID: 38385147 PMCID: PMC10879280 DOI: 10.3389/fncel.2024.1337339] [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: 11/12/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
B cells are fundamental players in the pathophysiology of autoimmune diseases of the central nervous system, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). A deeper understanding of disease-specific B cell functions has led to the differentiation of both diseases and the development of different treatment strategies. While NMOSD is strongly associated with pathogenic anti-AQP4 IgG antibodies and proinflammatory cytokine pathways, no valid autoantibodies have been identified in MS yet, apart from certain antigen targets that require further evaluation. Although both diseases can be effectively treated with B cell depleting therapies, there are distinct differences in the peripheral B cell subsets that influence CNS inflammation. An increased peripheral blood double negative B cells (DN B cells) and plasmablast populations has been demonstrated in NMOSD, but not consistently in MS patients. Furthermore, DN B cells are also elevated in rheumatic diseases and other autoimmune entities such as myasthenia gravis and Guillain-Barré syndrome, providing indirect evidence for a possible involvement of DN B cells in other autoantibody-mediated diseases. In MS, the peripheral memory B cell pool is affected by many treatments, providing indirect evidence for the involvement of memory B cells in MS pathophysiology. Moreover, it must be considered that an important effector function of B cells in MS may be the presentation of antigens to peripheral immune cells, including T cells, since B cells have been shown to be able to recirculate in the periphery after encountering CNS antigens. In conclusion, there are clear differences in the composition of B cell populations in MS and NMOSD and treatment strategies differ, with the exception of broad B cell depletion. This review provides a detailed overview of the role of different B cell subsets in MS and NMOSD and their implications for treatment options. Specifically targeting DN B cells and plasmablasts in NMOSD as opposed to memory B cells in MS may result in more precise B cell therapies for both diseases.
Collapse
Affiliation(s)
| | | | | | - M. C. Kowarik
- Department of Neurology and Stroke, Center for Neurology, and Hertie-Institute for Clinical Brain Research Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Aspden JW, Murphy MA, Kashlan RD, Xiong Y, Poznansky MC, Sîrbulescu RF. Intruders or protectors - the multifaceted role of B cells in CNS disorders. Front Cell Neurosci 2024; 17:1329823. [PMID: 38269112 PMCID: PMC10806081 DOI: 10.3389/fncel.2023.1329823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
B lymphocytes are immune cells studied predominantly in the context of peripheral humoral immune responses against pathogens. Evidence has been accumulating in recent years on the diversity of immunomodulatory functions that B cells undertake, with particular relevance for pathologies of the central nervous system (CNS). This review summarizes current knowledge on B cell populations, localization, infiltration mechanisms, and function in the CNS and associated tissues. Acute and chronic neurodegenerative pathologies are examined in order to explore the complex, and sometimes conflicting, effects that B cells can have in each context, with implications for disease progression and treatment outcomes. Additional factors such as aging modulate the proportions and function of B cell subpopulations over time and are also discussed in the context of neuroinflammatory response and disease susceptibility. A better understanding of the multifactorial role of B cell populations in the CNS may ultimately lead to innovative therapeutic strategies for a variety of neurological conditions.
Collapse
Affiliation(s)
- James W. Aspden
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Matthew A. Murphy
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rommi D. Kashlan
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yueyue Xiong
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Ruxandra F. Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Neziraj T, Siewert L, Pössnecker E, Pröbstel AK. Therapeutic targeting of gut-originating regulatory B cells in neuroinflammatory diseases. Eur J Immunol 2023; 53:e2250033. [PMID: 37624875 DOI: 10.1002/eji.202250033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/29/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
Regulatory B cells (Bregs) are immunosuppressive cells that support immunological tolerance by the production of IL-10, IL-35, and TGF-β. Bregs arise from different developmental stages in response to inflammatory stimuli. In that regard, mounting evidence points towards a direct influence of gut microbiota on mucosal B cell development, activation, and regulation in health and disease. While an increasing number of diseases are associated with alterations in gut microbiome (dysbiosis), little is known about the role of microbiota on Breg development and induction in neuroinflammatory disorders. Notably, gut-originating, IL-10- and IgA-producing regulatory plasma cells have recently been demonstrated to egress from the gut to suppress inflammation in the CNS raising fundamental questions about the triggers and functions of mucosal-originating Bregs in systemic inflammation. Advancing our understanding of Bregs in neuroinflammatory diseases could lead to novel therapeutic approaches. Here, we summarize the main aspects of Breg differentiation and functions and evidence about their involvement in neuroinflammatory diseases. Further, we highlight current data of gut-originating Bregs and their microbial interactions and discuss future microbiota-regulatory B cell-targeted therapies in immune-mediated diseases.
Collapse
Affiliation(s)
- Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Lena Siewert
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Ma X, Ma R, Zhang M, Qian B, Wang B, Yang W. Recent Progress in Multiple Sclerosis Treatment Using Immune Cells as Targets. Pharmaceutics 2023; 15:pharmaceutics15030728. [PMID: 36986586 PMCID: PMC10057470 DOI: 10.3390/pharmaceutics15030728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-mediated demyelinating disease of the central nervous system. The main pathological features are inflammatory reaction, demyelination, axonal disintegration, reactive gliosis, etc. The etiology and pathogenesis of the disease have not been clarified. The initial studies believed that T cell-mediated cellular immunity is the key to the pathogenesis of MS. In recent years, more and more evidence has shown that B cells and their mediated humoral immune and innate immune cells (such as microglia, dendritic cells, macrophages, etc.) also play an important role in the pathogenesis of MS. This article mainly reviews the research progress of MS by targeting different immune cells and analyzes the action pathways of drugs. The types and mechanisms of immune cells related to the pathogenesis are introduced in detail, and the mechanisms of drugs targeting different immune cells are discussed in depth. This article aims to clarify the pathogenesis and immunotherapy pathway of MS, hoping to find new targets and strategies for the development of therapeutic drugs for MS.
Collapse
Affiliation(s)
- Xiaohong Ma
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Baicheng Qian
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Baoliang Wang
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- Correspondence: (B.W.); (W.Y.)
| | - Weijing Yang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (B.W.); (W.Y.)
| |
Collapse
|
6
|
Li Y, Zhang J, Liu L, Cui S, Sun H, Jiang H, Guo Y, Zhang J, Xie Z, Wang J. The imbalance between Bregs, Tfh, and Tregs in patients with anti-N-methyl-D-aspartate receptor encephalitis. Neurol Sci 2023:10.1007/s10072-023-06624-z. [PMID: 36781561 DOI: 10.1007/s10072-023-06624-z] [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: 11/19/2022] [Accepted: 01/12/2023] [Indexed: 02/15/2023]
Abstract
OBJECTIVE To detect the alteration of regulatory B cells (Bregs), follicular helper T cells (Tfh), and regulatory T cells (Tregs) frequencies in patients with anti-N-methyl-D-aspartate receptor (anti-NMDAR) encephalitis. Analyze their association with clinical severity and activity, and explore the effects of different immunotherapies on those immune cell subsets. METHODS We enrolled 21 patients with anti-NMDAR encephalitis, 22 patients with neuromyelitis optica spectrum disorder (NMOSD), 14 patients with idiopathic intracranial hypertension (IIH), and 20 healthy controls (HC) in our study. The frequencies of various immune cell subsets were determined using flow cytometry. RESULTS Compared to patients with IIH and HC, the frequencies of CD24hiCD38hi transitional B cells as well as Tregs were significantly lower while the frequency of Tfh was significantly higher in patients with anti-NMDAR encephalitis. The frequency of CD24hiCD38hi transitional B cells was significantly lower in the acute stage than in the recovery stage, and was negatively correlated with the modified Rankin scale (mRS) and the clinical assessment scale for autoimmune encephalitis (CASE). The frequency of CD24hiCD38hi transitional B cells at the last follow-up after rituximab (RTX) treatment was significantly higher than those treated with oral immunosuppressants or untreated. There was no clear difference between anti-NMDAR encephalitis and NMOSD in the above immune cell subsets. CONCLUSION We suggested that the frequencies of CD24hiCD38hi transitional B cells and Tregs were decreased while the frequency of Tfh was increased in patients with anti-NMDAR encephalitis. CD24hiCD38hi transitional B cells frequency may be a potential indicator to estimate the disease activity and severity.
Collapse
Affiliation(s)
- Yatong Li
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jing Zhang
- Clinical Research Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lei Liu
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shilei Cui
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Houliang Sun
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hanqiu Jiang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yanjun Guo
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jingxiao Zhang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhuxiao Xie
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiawei Wang
- Department of Neurology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Giovannini D, Belbezier A, Baillet A, Bouillet L, Kawano M, Dumestre-Perard C, Clavarino G, Noble J, Pers JO, Sturm N, Huard B. Heterogeneity of antibody-secreting cells infiltrating autoimmune tissues. Front Immunol 2023; 14:1111366. [PMID: 36895558 PMCID: PMC9989216 DOI: 10.3389/fimmu.2023.1111366] [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: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
The humoral response is frequently dysfunctioning in autoimmunity with a frequent rise in total serum immunoglobulins, among which are found autoantibodies that may be pathogenic by themselves and/or propagate the inflammatory reaction. The infiltration of autoimmune tissues by antibody-secreting cells (ASCs) constitutes another dysfunction. The known high dependency of ASCs on the microenvironment to survive combined to the high diversity of infiltrated tissues implies that ASCs must adapt. Some tissues even within a single clinical autoimmune entity are devoid of infiltration. The latter means that either the tissue is not permissive or ASCs fail to adapt. The origin of infiltrated ASCs is also variable. Indeed, ASCs may be commonly generated in the secondary lymphoid organ draining the autoimmune tissue, and home at the inflammation site under the guidance of specific chemokines. Alternatively, ASCs may be generated locally, when ectopic germinal centers are formed in the autoimmune tissue. Alloimmune tissues with the example of kidney transplantation will also be discussed own to their high similarity with autoimmune tissues. It should also be noted that antibody production is not the only function of ASCs, since cells with regulatory functions have also been described. This article will review all the phenotypic variations indicative of tissue adaptation described so for at the level of ASC-infiltrating auto/alloimmune tissues. The aim is to potentially define tissue-specific molecular targets in ASCs to improve the specificity of future autoimmune treatments.
Collapse
Affiliation(s)
- Diane Giovannini
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Aude Belbezier
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Athan Baillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Rheumatology, Grenoble University Hospital, Grenoble, France
| | - Laurence Bouillet
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France.,Department of Internal Medicine, Grenoble University Hospital, Grenoble, France
| | - Mitsuhiro Kawano
- Department of Rheumatology, Kanazawa University Hospital, Kanazawa, Japan
| | | | | | - Johan Noble
- Department of Nephrology, Grenoble University Hospital, Grenoble, France
| | - Jacques-Olivier Pers
- B Lymphocytes, Autoimmunity and Immunotherapies, Brest University, INSERM, UMR1227, Brest, France.,Odontology Unit, Brest University Hospital, Brest, France
| | - Nathalie Sturm
- Department of Pathology, Grenoble University Hospital, Grenoble, France.,Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| | - Bertrand Huard
- Translational Research in Autoimmunity and Inflammation Group (TRAIG), Translational Innovation in Medicine and Complexity (TIMC), University Grenoble-Alpes, CNRS Unité mixte de recherche (UMR) 5525, Grenoble, France
| |
Collapse
|
8
|
Redenbaugh V, Flanagan EP. Monoclonal Antibody Therapies Beyond Complement for NMOSD and MOGAD. Neurotherapeutics 2022; 19:808-822. [PMID: 35267170 PMCID: PMC9294102 DOI: 10.1007/s13311-022-01206-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 01/09/2023] Open
Abstract
Aquaporin-4 (AQP4)-IgG seropositive neuromyelitis optica spectrum disorders (AQP4-IgG seropositive NMOSD) and myelin oligodendrocyte glycoprotein (MOG)-IgG-associated disease (MOGAD) are inflammatory demyelinating disorders distinct from each other and from multiple sclerosis (MS).While anti-CD20 treatments can be used to treat MS and AQP4-IgG seropositive NMOSD, some MS medications are ineffective or could exacerbate AQP4-IgG seropositive NMOSD including beta-interferons, natalizumab, and fingolimod. AQP4-IgG seropositive NMOSD has a relapsing course in most cases, and preventative maintenance treatments should be started after the initial attack. Rituximab, eculizumab, inebilizumab, and satralizumab all have class 1 evidence for use in AQP4-IgG seropositive NMOSD, and the latter three have been approved by the US Food and Drug Administration (FDA). MOGAD is much more likely to be monophasic than AQP4-IgG seropositive NMOSD, and preventative therapy is usually reserved for those who have had a disease relapse. There is a lack of any class 1 evidence for MOGAD preventative treatment. Observational benefit has been suggested from oral immunosuppressants, intravenous immunoglobulin (IVIg), rituximab, and tocilizumab. Randomized placebo-controlled trials are urgently needed in this area.
Collapse
Affiliation(s)
- Vyanka Redenbaugh
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Eoin P Flanagan
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| |
Collapse
|
9
|
Stathopoulos P, Dalakas MC. Evolution of Anti-B Cell Therapeutics in Autoimmune Neurological Diseases. Neurotherapeutics 2022; 19:691-710. [PMID: 35182380 PMCID: PMC9294112 DOI: 10.1007/s13311-022-01196-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
Collapse
Affiliation(s)
- Panos Stathopoulos
- 1st Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
10
|
Wang L, Huang W, ZhangBao J, Chang X, Tan H, Zhou L, Lu C, Wang M, Lu J, Zhao C, Quan C. The Alteration of Circulating Lymphocyte Subsets During Tacrolimus Therapy in Neuromyelitis Optica Spectrum Disorder and Its Correlation With Clinical Outcomes. Front Neurol 2022; 12:816721. [PMID: 35126303 PMCID: PMC8809081 DOI: 10.3389/fneur.2021.816721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
ObjectivesWe aimed to explore the alteration of circulating lymphocyte subsets before and after tacrolimus (TAC) therapy in neuromyelitis optica spectrum disorder (NMOSD) and its correlation with clinical outcomes.MethodsAnti-aquaporin-4 antibody (AQP4-ab)-positive patients with NMOSD treated with TAC were followed and clinically evaluated at 0, 3, 6, and 12 months after initiation of TAC. Flow cytometry was employed to detect the proportion of various whole blood lymphocyte subsets at every time point. Correlation analysis was further performed to explore the association between annualized relapse rate (ARR), the Expanded Disability Status Scale (EDSS) score, and the proportion of circulating lymphocyte subsets before and after TAC therapy.ResultsA total of 13 eligible patients with NMOSD were included. The proportion of CD19+CD24hiCD38hi/CD19+ and CD19+CD5+CD1dhi/CD19+ lymphocyte subsets increased significantly after TAC therapy (p = 0.010 and p < 0.001). The proportion of CD19+BAFFR+, CD19+IFN-γ+, and CD19+IL-10+ subsets decreased significantly after TAC therapy (p = 0.015, 0.018, and 0.042, respectively). There was a negative correlation between CD4+CD25hi subset and EDSS score (p = 0.016, r = −0.652).ConclusionPossibly through increasing regulatory B and suppressing BAFFR+ B and interferon (IFN)-γ+ B subsets, TAC could decrease relapse. EDSS score may be correlated with some lymphocyte subsets after TAC therapy.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Wenjuan Huang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Jingzi ZhangBao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Xuechun Chang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Hongmei Tan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Lei Zhou
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Chuanzhen Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Min Wang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
| | - Chao Quan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders (NCND), Shanghai, China
- *Correspondence: Chao Quan
| |
Collapse
|
11
|
Kim Y, Kim SY, Han SM, Payumo RM, Park K, Kim HE, Kim SH, Hyun JW, Lee E, Kim HJ. Functional impairment of CD19 +CD24 hiCD38 hi B cells in neuromyelitis optica spectrum disorder is restored by B cell depletion therapy. Sci Transl Med 2021; 13:eabk2132. [PMID: 34910550 DOI: 10.1126/scitranslmed.abk2132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yeseul Kim
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea.,Yonsei University College of Medicine, Seoul 03772, Korea.,Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - So Yeon Kim
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea.,Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Sang-Min Han
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea.,Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Rosah May Payumo
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Kevin Park
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea.,Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Ha Eun Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Su-Hyun Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Jae-Won Hyun
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| | - Eunjig Lee
- Yonsei University College of Medicine, Seoul 03772, Korea
| | - Ho Jin Kim
- Division of Clinical Research, Research Institute, National Cancer Center, Goyang 10408, Korea.,Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang 10408, Korea
| |
Collapse
|
12
|
Ghafouri-Fard S, Azimi T, Taheri M. A Comprehensive Review on the Role of Genetic Factors in Neuromyelitis Optica Spectrum Disorder. Front Immunol 2021; 12:737673. [PMID: 34675927 PMCID: PMC8524039 DOI: 10.3389/fimmu.2021.737673] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) comprise a variety of disorders being described by optic neuritis and myelitis. This disorder is mostly observed in sporadic form, yet 3% of cases are familial NMO. Different series of familial NMO cases have been reported up to now, with some of them being associated with certain HLA haplotypes. Assessment of HLA allele and haplotypes has also revealed association between some alleles within HLA-DRB1 or other loci and sporadic NMO. More recently, genome-wide SNP arrays have shown some susceptibility loci for NMO. In the current manuscript, we review available information about the role of genetic factors in NMO.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Azimi
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakin Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Carnero Contentti E, Correale J. Neuromyelitis optica spectrum disorders: from pathophysiology to therapeutic strategies. J Neuroinflammation 2021; 18:208. [PMID: 34530847 PMCID: PMC8444436 DOI: 10.1186/s12974-021-02249-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023] Open
Abstract
Neuromyelitis optica (NMO) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) characterized by acute optic neuritis (ON) and transverse myelitis (TM). NMO is caused by a pathogenic serum IgG antibody against the water channel aquoporin 4 (AQP4) in the majority of patients. AQP4-antibody (AQP4-ab) presence is highly specific, and differentiates NMO from multiple sclerosis. It binds to AQP4 channels on astrocytes, triggering activation of the classical complement cascade, causing granulocyte, eosinophil, and lymphocyte infiltration, culminating in injury first to astrocyte, then oligodendrocytes followed by demyelination and neuronal loss. NMO spectrum disorder (NMOSD) has recently been defined and stratified based on AQP4-ab serology status. Most NMOSD patients experience severe relapses leading to permanent neurologic disability, making suppression of relapse frequency and severity, the primary objective in disease management. The most common treatments used for relapses are steroids and plasma exchange.Currently, long-term NMOSD relapse prevention includes off-label use of immunosuppressants, particularly rituximab. In the last 2 years however, three pivotal clinical trials have expanded the spectrum of drugs available for NMOSD patients. Phase III studies have shown significant relapse reduction compared to placebo in AQP4-ab-positive patients treated with satralizumab, an interleukin-6 receptor (IL-6R) inhibitor, inebilizumab, an antibody against CD19+ B cells; and eculizumab, an antibody blocking the C5 component of complement. In light of the new evidence on NMOSD pathophysiology and of preliminary results from ongoing trials with new drugs, we present this descriptive review, highlighting promising treatment modalities as well as auspicious preclinical and clinical studies.
Collapse
|
14
|
Treatment of Neuromyelitis Optica Spectrum Disorders. Int J Mol Sci 2021; 22:ijms22168638. [PMID: 34445343 PMCID: PMC8395403 DOI: 10.3390/ijms22168638] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune central nervous system (CNS) inflammatory disorder that can lead to serious disability and mortality. Females are predominantly affected, including those within the reproductive age. Most patients develop relapsing attacks of optic neuritis; longitudinally extensive transverse myelitis; and encephalitis, especially brainstem encephalitis. The majority of NMOSD patients are seropositive for IgG autoantibodies against the water channel protein aquaporin-4 (AQP4-IgG), reflecting underlying aquaporin-4 autoimmunity. Histological findings of the affected CNS tissues of patients from in-vitro and in-vivo studies support that AQP4-IgG is directly pathogenic in NMOSD. It is believed that the binding of AQP4-IgG to CNS aquaporin-4 (abundantly expressed at the endfoot processes of astrocytes) triggers astrocytopathy and neuroinflammation, resulting in acute attacks. These attacks of neuroinflammation can lead to pathologies, including aquaporin-4 loss, astrocytic activation, injury and loss, glutamate excitotoxicity, microglial activation, neuroinflammation, demyelination, and neuronal injury, via both complement-dependent and complement-independent pathophysiological mechanisms. With the increased understanding of these mechanisms underlying this serious autoimmune astrocytopathy, effective treatments for both active attacks and long-term immunosuppression to prevent relapses in NMOSD are increasingly available based on the evidence from retrospective observational data and prospective clinical trials. Knowledge on the indications and potential side effects of these medications are essential for a clear evaluation of the potential benefits and risks to NMOSD patients in a personalized manner. Special issues such as pregnancy and the coexistence of other autoimmune diseases require additional concern and meticulous care. Future directions include the identification of clinically useful biomarkers for the prediction of relapse and monitoring of the therapeutic response, as well as the development of effective medications with minimal side effects, especially opportunistic infections complicated by long-term immunosuppression.
Collapse
|
15
|
Wu H, Chen X, Gu F, Zhang P, Xu S, Liu Q, Zhang Q, Wang X, Wang C, Körner H, Wei W. CP-25 alleviates antigen-induced experimental Sjögren's syndrome in mice by inhibiting JAK1-STAT1/2-CXCL13 signaling and interfering with B-cell migration. J Transl Med 2021; 101:1084-1097. [PMID: 32620868 DOI: 10.1038/s41374-020-0453-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 06/02/2020] [Accepted: 06/02/2020] [Indexed: 02/08/2023] Open
Abstract
The etiology of primary Sjögren's syndrome (pSS) remains unknown, and there is no complete curative drug. In this study, we treated a mouse model of the submandibular gland (SG) protein-immunized experimental Sjögren's syndrome (ESS) with paeoniflorin-6'-O-benzene sulfonate (termed CP-25) to evaluate the potential therapeutic effects of CP-25. Through in vivo experiments, we found that CP-25 increased saliva flow, alleviated the salivary gland indexes, and improved tissue integrity in the ESS model. The viability of splenocytes and B-lymphocyte migration from spleen were reduced in ESS mice. Furthermore, CP-25 decreased the expression of IgG antibodies, anti-SSA and anti-SSB antibodies and modulated the levels of cytokines in the serum of SS mice. The numbers of total B lymphocytes, plasma cells (PCs), and memory B cells diminished in the salivary gland. Increased expression of the JAK1-STAT1-CXCL13 axis and IFNα was found in human tissue isolated from pSS patients. In vitro, after stimulation with IFNα, the levels of CXCL13 mRNA and CXCL13 in human salivary gland epithelial cells (HSGEC) increased, while CP-25 counteracted the secretion of CXCL13 and downregulated the expression of CXCL13. IFN-α activated the JAK1-STAT1/2-CXCL13 signaling pathway in HSGEC, which was negatively regulated by additional CP-25. As a consequence, B-cell migration was downregulated in coculture with IFN-α-stimulated HSGEC. Taken together, this study demonstrated that the therapeutic effects of CP-25 were associated with the inhibition of the JAK1-STAT1/2-CXCL13 signaling pathway in HSGEC, which impedes the migration of B cells into the salivary gland. We identified the underlying mechanisms of the therapeutic effect of CP-25 and provided an experimental foundation for CP-25 as a potential drug in the treatment of the human autoimmune disorder pSS.
Collapse
Affiliation(s)
- Huaxun Wu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China.
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China.
| | - Xiaoyun Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Fang Gu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Pengying Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Shixia Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Qi Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Qiaolin Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Xinming Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Heinrich Körner
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, 230032, Anhui, China.
- Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032, Anhui, China.
| |
Collapse
|
16
|
Xiang W, Xie C, Guan Y. The identification, development and therapeutic potential of IL-10-producing regulatory B cells in multiple sclerosis. J Neuroimmunol 2021; 354:577520. [PMID: 33684831 DOI: 10.1016/j.jneuroim.2021.577520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/27/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
Regulatory B cells are a rare B-cell subset widely known to exert their immunosuppressive function via the production of interleukin-10 (IL-10) and other mechanisms. B10 cells are a special subset of regulatory B cells with immunoregulatory function that is fully attributed to IL-10. Their unique roles in the animal model of multiple sclerosis (MS) have been described, as well as their relevance in MS patients. This review specifically focuses on the identification and development of B10 cells, the signals that promote IL-10 production in B cells, the roles of B10 cells in MS, and the potential and major challenges of the application of B10-based therapies for MS.
Collapse
Affiliation(s)
- Weiwei Xiang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Rd, Shanghai 200127, China
| | - Chong Xie
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Rd, Shanghai 200127, China
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Rd, Shanghai 200127, China.
| |
Collapse
|
17
|
Increased Frequencies of Switched Memory B Cells and Plasmablasts in Peripheral Blood from Patients with ANCA-Associated Vasculitis. J Immunol Res 2020; 2020:8209737. [PMID: 33313327 PMCID: PMC7719539 DOI: 10.1155/2020/8209737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/23/2020] [Accepted: 11/04/2020] [Indexed: 11/17/2022] Open
Abstract
B cells are thought to play a central role in the pathogenesis of antineutrophil cytoplasmic antibody- (ANCA-) associated vasculitis (AAV). ANCAs have been proposed to cause vasculitis by activating primed neutrophils to damage small blood vessels. We studied a cohort of AAV patients of which a majority were in remission and diagnosed with granulomatosis with polyangiitis (GPA). Using flow cytometry, the frequencies of CD19+ B cells and subsets in peripheral blood from 106 patients with AAV and 134 healthy controls were assessed. B cells were divided into naive, preswitch memory, switched memory, and exhausted memory cells. Naive and switched memory cells were further subdivided into transitional cells and plasmablasts, respectively. In addition, serum concentrations of immunoglobulin A, G, and M were measured and clinical data were retrieved. AAV patients displayed, in relation to healthy controls, a decreased frequency of B cells of lymphocytes (5.1% vs. 8.3%) and total B cell number. For the subsets, a decrease in percentage of transitional B cells (0.7% vs. 4.4%) and expansions of switched memory B cells (22.3% vs. 16.5%) and plasmablasts (0.9% vs. 0.3%) were seen. A higher proportion of B cells was activated (CD95+) in patients (20.6% vs. 10.3%), and immunoglobulin levels were largely unaltered. No differences in B cell frequencies between patients in active disease and remission were observed. Patients in remission with a tendency to relapse had, compared to nonrelapsing patients, decreased frequencies of B cells (3.5% vs. 6.5%) and transitional B cells (0.1% vs. 1.1%) and an increased frequency of activated exhausted memory B cells (30.8% vs. 22.3%). AAV patients exhibit specific changes in frequencies of CD19+ B cells and their subsets in peripheral blood. These alterations could contribute to the autoantibody-driven inflammatory process in AAV.
Collapse
|
18
|
Eslami M, Meinl E, Eibel H, Willen L, Donzé O, Distl O, Schneider H, Speiser DE, Tsiantoulas D, Yalkinoglu Ö, Samy E, Schneider P. BAFF 60-mer, and Differential BAFF 60-mer Dissociating Activities in Human Serum, Cord Blood and Cerebrospinal Fluid. Front Cell Dev Biol 2020; 8:577662. [PMID: 33240880 PMCID: PMC7677505 DOI: 10.3389/fcell.2020.577662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
B cell activation factor of the TNF family (BAFF/BLyS), an essential B cell survival factor of which circulating levels are elevated in several autoimmune disorders, is targeted in the clinic for the treatment of systemic lupus erythematosus (SLE). The soluble form of BAFF can exist as 3-mer, or as 60-mer that results from the ordered assembly of twenty 3-mers and that can be obtained from naturally cleaved membrane-bound BAFF or made as a recombinant protein. However, which forms of soluble BAFF exist and act in humans is unclear. In this study, BAFF 3-mer and 60-mer in biological fluids were characterized for size, activity and response to specific stimulators or inhibitors of BAFF. Human cerebrospinal fluids (CSF) from patients with multiple sclerosis and adult human sera contained exclusively BAFF 3-mer in these assays, also when BAFF concentrations were moderately SLE or highly (BAFFR-deficient individual) increased. Human sera, but not CSF, contained a high molecular weight, saturable activity that dissociated preformed recombinant BAFF 60-mer into 3-mer. This activity was lower in cord blood. Cord blood displayed BAFF levels 10-fold higher than in adults and consistently contained a fair proportion of active high molecular weight BAFF able to dissociate into 3-mer but not endowed with all properties of recombinant BAFF 60-mer. If BAFF 60-mer is produced in humans, it is dissociated, or at least attenuated in the circulation.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital of the Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann Eibel
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Freiburg, Germany
| | - Laure Willen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Holm Schneider
- Department of Pediatrics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel E Speiser
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | | | - Özkan Yalkinoglu
- Clinical Pharmacology, Quantitative Pharmacology, Translational Medicine, Merck KGaA, Darmstadt, Germany
| | - Eileen Samy
- Business of Merck KGaA, EMD Serono Research & Development Institute, Inc., Billerica, MA, United States
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
19
|
Jarius S, Paul F, Weinshenker BG, Levy M, Kim HJ, Wildemann B. Neuromyelitis optica. Nat Rev Dis Primers 2020; 6:85. [PMID: 33093467 DOI: 10.1038/s41572-020-0214-9] [Citation(s) in RCA: 244] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Neuromyelitis optica (NMO; also known as Devic syndrome) is a clinical syndrome characterized by attacks of acute optic neuritis and transverse myelitis. In most patients, NMO is caused by pathogenetic serum IgG autoantibodies to aquaporin 4 (AQP4), the most abundant water-channel protein in the central nervous system. In a subset of patients negative for AQP4-IgG, pathogenetic serum IgG antibodies to myelin oligodendrocyte glycoprotein, an antigen in the outer myelin sheath of central nervous system neurons, are present. Other causes of NMO (such as paraneoplastic disorders and neurosarcoidosis) are rare. NMO was previously associated with a poor prognosis; however, treatment with steroids and plasma exchange for acute attacks and with immunosuppressants (in particular, B cell-depleting agents) for attack prevention has greatly improved the long-term outcomes. Recently, a number of randomized controlled trials have been completed and the first drugs, all therapeutic monoclonal antibodies, have been approved for the treatment of AQP4-IgG-positive NMO and its formes frustes.
Collapse
Affiliation(s)
- Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Thoman ME, McKarns SC. Metabolomic Profiling in Neuromyelitis Optica Spectrum Disorder Biomarker Discovery. Metabolites 2020; 10:metabo10090374. [PMID: 32961928 PMCID: PMC7570337 DOI: 10.3390/metabo10090374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
There is no specific test for diagnosing neuromyelitis optica spectrum disorder (NMOSD), a disabling autoimmune disease of the central nervous system. Instead, diagnosis relies on ruling out other related disorders with overlapping clinical symptoms. An urgency for NMOSD biomarker discovery is underscored by adverse responses to treatment following misdiagnosis and poor prognosis following the delayed onset of treatment. Pathogenic autoantibiotics that target the water channel aquaporin-4 (AQP4) and myelin oligodendrocyte glycoprotein (MOG) contribute to NMOSD pathology. The importance of early diagnosis between AQP4-Ab+ NMOSD, MOG-Ab+ NMOSD, AQP4-Ab− MOG-Ab− NMOSD, and related disorders cannot be overemphasized. Here, we provide a comprehensive data collection and analysis of the currently known metabolomic perturbations and related proteomic outcomes of NMOSD. We highlight short chain fatty acids, lipoproteins, amino acids, and lactate as candidate diagnostic biomarkers. Although the application of metabolomic profiling to individual NMOSD patient care shows promise, more research is needed.
Collapse
Affiliation(s)
- Maxton E. Thoman
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Susan C. McKarns
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Correspondence:
| |
Collapse
|
21
|
Klein da Costa B, Brant de Souza Melo R, Passos GRD, Gomes Meneses Sevilha Castro D, Becker J, Bar-Or A, Sato DK. Unraveling B lymphocytes in CNS inflammatory diseases: Distinct mechanisms and treatment targets. Neurology 2020; 95:733-744. [PMID: 32907966 DOI: 10.1212/wnl.0000000000010789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
Specific therapies targeting B lymphocytes in multiple sclerosis (MS) have demonstrated reductions in disease activity and disability progression. Several observational studies have also shown the effects of targeting B lymphocytes in other rare CNS inflammatory diseases, such as neuromyelitis optica spectrum disorder (NMOSD) and autoimmune encephalitis (AE). However, some drugs targeting cytokine receptors involved in B-lymphocyte maturation and proliferation resulted in negative outcomes in MS. These apparently conflicting findings have stimulated research on the pathophysiologic mechanisms of B lymphocytes in CNS inflammatory diseases. It has been demonstrated that B lymphocytes participate in the pathogenesis of these conditions as antigen-presenting cells, producing proinflammatory cytokines that induce Th1 and Th17 responses and producing antibodies. However, they are also able to produce anti-inflammatory cytokines, such as interleukin-10, functioning as regulators of autoimmunity. Understanding these diverse effects is essential for the development of focused treatments. In this review, we discuss the possible mechanisms that underlie B-lymphocyte involvement in MS, NMOSD, and AE and the outcomes obtained by treatments targeting B lymphocytes.
Collapse
Affiliation(s)
- Bruna Klein da Costa
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia.
| | - Renata Brant de Souza Melo
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Giordani Rodrigues Dos Passos
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Douglas Gomes Meneses Sevilha Castro
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Jefferson Becker
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Amit Bar-Or
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| | - Douglas Kazutoshi Sato
- From the Brain Institute of Rio Grande do Sul (BraIns) and School of Medicine (B.K.d.C., R.B.d.S.M., G.R.d.P., J.B., D.K.S.), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre; Santa Casa de Belo Horizonte (R.B.d.S.M.), Belo Horizonte; Produtos Roche Químicos e Farmacêuticos S.A. (D.G.M.S.C.), São Paulo, Brazil; and Perelman Center for Advanced Medicine (PCAM) (A.B.-O), University of Pennsylvania, Philadelphia
| |
Collapse
|
22
|
Yang MG, Tian S, Zhang Q, Han J, Liu C, Zhou Y, Zhu J, Jin T. Elevated serum interleukin-39 levels in patients with neuromyelitis optica spectrum disorders correlated with disease severity. Mult Scler Relat Disord 2020; 46:102430. [PMID: 32853892 DOI: 10.1016/j.msard.2020.102430] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/17/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023]
Abstract
AIMS Neuromyelitis optica spectrum disorders (NMOSD) is an inflammatory demyelinating autoimmune disorder in the central nervous system (CNS), which is mainly mediated by aquaporin 4 antibodies (AQP4-Ab). Interleukin (IL)-39 is a new pro-inflammatory cytokine which belongs to the IL-12 cytokine family. However, the role of IL-39 in patients with NMOSD is not completely understood. Therefore, the aim of this study is to explore the possible implication of IL-39 in the pathogenesis of NMOSD. METHODS In this study, 50 patients with NMOSD, 20 patients with relapsing-remitting multiple sclerosis (RRMS), 30 patients with non-inflammatory neurological disorders (NND) and 78 healthy controls (HCs) were recruited. The levels of serum IL-39 were measured using the enzyme-linked immunosorbent assay (ELISA). RESULTS Our study showed serum IL-39 levels in patients with NMOSD were significantly higher than that in RRMS patients, NND patients and HCs, and positively correlated with Expanded Disability Status Scale (EDSS) score. CONCLUSION These findings suggested that IL-39 may play a pro-inflammatory role in the pathogenesis of NMOSD and correlate with disease severity.
Collapse
Affiliation(s)
- Meng-Ge Yang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Suyan Tian
- Division of Clinical Research, the First Hospital of Jilin University, Changchun, China
| | - Qingxiang Zhang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Caiyun Liu
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Yang Zhou
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
23
|
Ran Z, Yue-Bei L, Qiu-Ming Z, Huan Y. Regulatory B Cells and Its Role in Central Nervous System Inflammatory Demyelinating Diseases. Front Immunol 2020; 11:1884. [PMID: 32973780 PMCID: PMC7468432 DOI: 10.3389/fimmu.2020.01884] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Regulatory B (Breg) cells represent a population of suppressor B cells that participate in immunomodulatory processes and inhibition of excessive inflammation. The regulatory function of Breg cells have been demonstrated in mice and human with inflammatory diseases, cancer, after transplantation, and particularly in autoinflammatory disorders. In order to suppress inflammation, Breg cells produce anti-inflammatory mediators, induce death ligand-mediated apoptosis, and regulate many kinds of immune cells such as suppressing the proliferation and differentiation of effector T cell and increasing the number of regulatory T cells. Central nervous system Inflammatory demyelinating diseases (CNS IDDs) are a heterogeneous group of disorders, which occur against the background of an acute or chronic inflammatory process. With the advent of monoclonal antibodies directed against B cells, breakthroughs have been made in the treatment of CNS IDDs. Therefore, the number and function of B cells in IDDs have attracted attention. Meanwhile, increasing number of studies have confirmed that Breg cells play a role in alleviating autoimmune diseases, and treatment with Breg cells has also been proposed as a new therapeutic direction. In this review, we focus on the understanding of the development and function of Breg cells and on the diversification of Breg cells in CNS IDDs.
Collapse
Affiliation(s)
- Zhou Ran
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Luo Yue-Bei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeng Qiu-Ming
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Huan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Differential Effects of MS Therapeutics on B Cells-Implications for Their Use and Failure in AQP4-Positive NMOSD Patients. Int J Mol Sci 2020; 21:ijms21145021. [PMID: 32708663 PMCID: PMC7404039 DOI: 10.3390/ijms21145021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/25/2022] Open
Abstract
B cells are considered major contributors to multiple sclerosis (MS) pathophysiology. While lately approved disease-modifying drugs like ocrelizumab deplete B cells directly, most MS medications were not primarily designed to target B cells. Here, we review the current understanding how approved MS medications affect peripheral B lymphocytes in humans. These highly contrasting effects are of substantial importance when considering these drugs as therapy for neuromyelitis optica spectrum disorders (NMOSD), a frequent differential diagnosis to MS, which is considered being a primarily B cell- and antibody-driven diseases. Data indicates that MS medications, which deplete B cells or induce an anti-inflammatory phenotype of the remaining ones, were effective and safe in aquaporin-4 antibody positive NMOSD. In contrast, drugs such as natalizumab and interferon-β, which lead to activation and accumulation of B cells in the peripheral blood, lack efficacy or even induce catastrophic disease activity in NMOSD. Hence, we conclude that the differential effect of MS drugs on B cells is one potential parameter determining the therapeutic efficacy or failure in antibody-dependent diseases like seropositive NMOSD.
Collapse
|
25
|
Zhou Y, Zhang Y, Han J, Yang M, Zhu J, Jin T. Transitional B cells involved in autoimmunity and their impact on neuroimmunological diseases. J Transl Med 2020; 18:131. [PMID: 32183811 PMCID: PMC7079408 DOI: 10.1186/s12967-020-02289-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/28/2020] [Indexed: 02/08/2023] Open
Abstract
Transitional B cells (TrB cells) represent a crucial link between immature B cells in the bone marrow and mature peripheral B cells. Although TrB cells represent one of the regulatory B cell subpopulations in healthy individuals, the frequency of CD24hiCD38hi TrB cells in circulation may be altered in individuals with autoimmune diseases, such as multiple sclerosis, neuromyelitisoptica spectrum disorders, systemic lupus erythematosus, Sjögren’s syndrome, rheumatoid arthritis, systemic sclerosis, and juvenile dermatomyositis. Although TrB cells play regulatory roles under inflammatory conditions, consequences of their functional impairment vary across autoimmune diseases. Since the origin, development, and function of TrB cells, especially in humans, remain unclear and controversial, this review aimed to discuss the characteristics of TrB cells at steady state and explore their role in various immune diseases, including autoimmune rheumatic diseases and neuroimmunological diseases.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Mengge Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
26
|
Tradtrantip L, Asavapanumas N, Verkman AS. Emerging therapeutic targets for neuromyelitis optica spectrum disorder. Expert Opin Ther Targets 2020; 24:219-229. [PMID: 32070155 DOI: 10.1080/14728222.2020.1732927] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelinating disease of the central nervous system affecting primarily the spinal cord and optic nerves. Most NMOSD patients are seropositive for immunoglobulin G autoantibodies against astrocyte water channel aquaporin-4, called AQP4-IgG, which cause astrocyte injury leading to demyelination and neurological impairment. Current therapy for AQP4-IgG seropositive NMOSD includes immunosuppression, B cell depletion, and plasma exchange. Newer therapies target complement, CD19 and IL-6 receptors.Areas covered: This review covers early-stage pre-clinical therapeutic approaches for seropositive NMOSD. Targets include pathogenic AQP4-IgG autoantibodies and their binding to AQP4, complement-dependent and cell-mediated cytotoxicity, blood-brain barrier, remyelination and immune effector and regulatory cells, with treatment modalities including small molecules, biologics, and cells.Expert opinion: Though newer NMOSD therapies appear to have increased efficacy in reducing relapse rate and neurological deficit, increasingly targeted therapies could benefit NMOSD patients with ongoing relapses and could potentially be superior in efficacy and safety. Of the various early-stage therapeutic approaches, IgG inactivating enzymes, aquaporumab blocking antibodies, drugs targeting early components of the classical complement system, complement regulator-targeted drugs, and Fc-based multimers are of interest. Curative strategies, perhaps involving AQP4 tolerization, remain intriguing future possibilities.
Collapse
Affiliation(s)
- Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
27
|
Li X, Wang L, Zhou L, ZhangBao J, Miao MZ, Lu C, Lu J, Quan C. The imbalance between regulatory and memory B cells accompanied by an increased number of circulating T-follicular helper cells in MOG-antibody-associated demyelination. Mult Scler Relat Disord 2019; 36:101397. [PMID: 31546225 DOI: 10.1016/j.msard.2019.101397] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/25/2019] [Accepted: 09/15/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To explore the alteration of T and B lymphocyte subsets proportions in myelin oligodendrocyte glycoprotein (MOG)-antibody-associated demyelination. METHODS 19 MOG-antibody-positive, 25 AQP4-antibody-positive and 25 double-negative NMOSD patients in the acute phase of the diseases were included in the study, as well as 29 healthy controls. The frequencies of different lymphocyte subsets, including CD19+CD27+ memory B cells, CD19+CD24hiCD38hi, and CD19+CD5+CD1dhi regulatory B cells, IFN-γexpressing B cells, IL-10 expressing B cells and CD4+CXCR5+ICOS+T-follicular helper cells (TFH) were measured via flow cytometry and compared among the four groups. RESULTS The frequencies of CD19+CD24hiCD38hi, CD19+CD5+CD1dhi regulatory B cells as well as the IL-10 expressing B cells were significantly lower in the MOG-antibody-associated demyelination compared to the healthy controls, whereas the frequencies of CD19+CD27+ memory B cells were significantly higher in the MOG-antibody-positive group. The frequencies of TFH were significantly higher in the MOG-antibody-positive group as compared to the healthy controls. No significant difference was detected in the above mentioned lymphocytic profile between the MOG-antibody-positive and the AQP4-antibody-positive groups. CONCLUSIONS The immuno-regulatory functions of B cells were significantly impaired whereas TFH cells were markedly increased in the acute phase of MOG-antibody-associated demyelination. Despite having distinct clinical features, MOG-antibody-associated demyelination shared a similar lymphocytic profile with AQP4-antibody-positive NMOSD in the acute relapse phase.
Collapse
Affiliation(s)
- Xiaoyang Li
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China; Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Lei Zhou
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jingzi ZhangBao
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Michael Z Miao
- Department of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, United States; Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Chuanzhen Lu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China
| | - Chao Quan
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, 12 Wulumuqi Zhong Road, Shanghai 200040, China.
| |
Collapse
|
28
|
Disease course, progression and activity of neuromyelitis optica (NMOSD) in patients who were treated with Rituximab, 6 and 12 months after receiving the first dose of drug, in Isfahan city. Mult Scler Relat Disord 2019; 34:77-82. [DOI: 10.1016/j.msard.2019.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/08/2019] [Accepted: 06/15/2019] [Indexed: 11/21/2022]
|
29
|
Wu Y, Zhong L, Geng J. Neuromyelitis optica spectrum disorder: Pathogenesis, treatment, and experimental models. Mult Scler Relat Disord 2018; 27:412-418. [PMID: 30530071 DOI: 10.1016/j.msard.2018.12.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/21/2018] [Accepted: 12/02/2018] [Indexed: 01/10/2023]
Abstract
Neuromyelitis optica (NMO) and NMO spectrum disorder (NMOSD) are inflammatory CNS syndromes mainly involving the optic nerve and/or spinal cord and characterized by the presence of serum aquaporin-4 immunoglobulin G antibodies (AQP4-IgG). The pathology of NMOSD is complicated, while therapies for NMOSD are limited and only partially effective in most cases. This review article focuses on the main pathology of NMOSD involving AQP4-IgG and lymphocyte function. We also review the existing therapeutic methods and potential new treatments. Experimental NMO animal models are crucial for further research into NMO pathology and treatment. However, no AQP4-IgG-immunized animals have been reported. The establishment of NMO models is therefore difficult and primarily depends on the generation of transgenic mice or transcranial manipulation using human or monoclonal mouse anti-AQP4 antibodies. Advantages and disadvantages of each model are discussed.
Collapse
Key Words
- APC, antigen-presenting cell
- Abbreviations: ADCC, antibody-dependent cellular cytotoxicity
- Aqp4, aquaporin 4
- Aquaporin-4
- BAFF, b-cell activating factor
- BBB, blood-brain barrier
- BCR, b cell receptor
- CDD, complement-dependent cytotoxicity
- CFA, complete freund's adjuvant
- CSF, cerebrospinal fluid
- CXCL, c-x-c motif chemokine ligand
- EAE, experimental autoimmune encephalomyelitis
- ECD, extracellular domain
- Experimental animal models
- IGG, immunoglobulin g
- IVMP, methylprednisolone pulse
- LETM, longitudinally extensive transverse myelitis
- MAB, monoclonal antibody
- MBP, myelin-binding protein
- MOG, myelin oligodendrocyte glycoprotein
- MOG-Ab, anti-MOG antibody
- NF-H, neurofilament heavy chain
- NMO, neuromyelitis optica
- NMO-IgG, NMO with serum AQP4-IgG
- NMOSD, NMO spectrum disorder
- Neuromyelitis optica
- Neuromyelitis optica spectrum disorder
- PB, plasmablast
- PP, plasmapheresis
- Remyelination
Collapse
Affiliation(s)
- Yan Wu
- Department of Neurology, Xichang Road No.295, Kunming 650000, China.
| | - Lianmei Zhong
- Department of Neurology, Xichang Road No.295, Kunming 650000, China
| | - Jia Geng
- Department of Neurology, Xichang Road No.295, Kunming 650000, China
| |
Collapse
|
30
|
Ding YG, Chen G, Li Q, Wen XF, Wei L, Yang HS. Frequency of IL-10-producing regulatory B cells associated with disease activity in thyroid-associated orbitopathy. Int J Ophthalmol 2018; 11:1458-1462. [PMID: 30225218 DOI: 10.18240/ijo.2018.09.05] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/19/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the association between IL-10-producing regulatory B (B10) cells and the clinical features of thyroid-associated orbitopathy (TAO). METHODS A total of 30 patients with TAO were recruited at Zhongshan Ophthalmic Center from May 2015 to December 2015. Peripheral blood mononuclear cells (PBMCs) were separated from blood samples of 30 TAO patients and 16 healthy controls and stimulated with CD40 ligand and CpG for 48h. The frequency of IL-10+ B cells was examined by flow cytometry and the correlation between the frequency of IL-10+ B cells and clinical features of TAO was analyzed by SPSS. RESULTS The frequency of IL-10+ B cells among CD19+ B cells in TAO patients was significantly lower than in healthy controls (TAO: 4.66%±1.88% vs healthy control: 6.82%±2.40%, P<0.01). The frequency of IL-10+ B cells showed a positive correlation with disease activity of TAO measured by Clinical Activity Score (CAS) (r=0.50, P<0.01), and became higher in TAO patients with family history of Graves' disease (GD) (P=0.04). CONCLUSION The decrease of the frequency of IL-10+ B cells in TAO patients indicates the deficiency of B10 cells in TAO, and the positive association with disease activity suggests its important role in TAO inflammation regulation.
Collapse
Affiliation(s)
- Yun-Gang Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China.,Qindao Ludong Eye Hospital, Qingdao 266600, Shandong Province, China
| | - Guo Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Qian Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China.,Ningxia Eye Hospital, People's Hospital of Ningxia Hui Autonomous Region, the First Clinical College of Northwest University for Nationalities, the Cooperative Teaching Hospital of North Minzu University, Yinchuan 750001, Ningxia Hui Autonomous Region, China
| | - Xiao-Feng Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Hua-Sheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
31
|
Shu Y, Zhang L, Chang Y, Li R, Sun X, Li J, Wu H, Yang Y, Peng L, Lu Z, Kermode AG, Qiu W. Association of serum Cystatin C with neuromyelitis optica spectrum disorders. Eur J Neurol 2018; 25:999-1002. [PMID: 29603501 DOI: 10.1111/ene.13646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/22/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Y. Shu
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - L. Zhang
- Department of Neurology; The Fifth Affiliated Hospital of Sun Yat-sen University; Zhuhai Guangdong China
| | - Y. Chang
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - R. Li
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - X. Sun
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - J. Li
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - H. Wu
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Y. Yang
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - L. Peng
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Z. Lu
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - A. G. Kermode
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
- Department of Neurology; Centre for Neuromuscular and Neurological Disorders; Sir Charles Gairdner Hospital; Queen Elizabeth II Medical Centre; University of Western Australia; Perth Australia
| | - W. Qiu
- Department of Neurology; The Third Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| |
Collapse
|
32
|
The IL-10-producing regulatory B cells (B10 cells) and regulatory T cell subsets in neuromyelitis optica spectrum disorder. Neurol Sci 2018; 39:1307-1308. [PMID: 29502269 DOI: 10.1007/s10072-018-3303-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/23/2018] [Indexed: 01/11/2023]
|
33
|
The IL-10-producing regulatory B cells (B10 cells) and regulatory T cell subsets in neuromyelitis optica spectrum disorder. Neurol Sci 2018; 39:543-549. [PMID: 29349658 DOI: 10.1007/s10072-018-3248-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/05/2018] [Indexed: 01/07/2023]
Abstract
B cells contribute to the pathogenesis of neuromyelitis optica (NMO) by producing Aquaporin 4-specific autoantibodies (AQP4-ab); on the other hand, there are certain B cells that suppress immune responses by producing regulatory cytokines, such as IL-10. In this study, we investigated the presence of IL-10-producing Breg cells among lymphocyte subsets. Twenty-two seropositive NMO spectrum disorder (NMOSD) patients (29 samples) and 13 healthy controls (HCs) (14 samples) were enrolled. All NMOSD patients have received one or more immunosuppressive drugs. The phenotype and frequency of B cell and T cell subsets in the peripheral blood were measured by flow cytometry. We defined Breg cells as IL-10-producing B (B10) cells, which are CD19+CD39+CD1d+IL-10+. The potential relations were evaluated between specific lymphocyte subsets and AQP4-ab intensity measured by the cell-based indirect immunofluorescence assay. The frequency of B10 cells was higher in patients with NMOSD regardless of the disease status than that in HCs (attack samples; p = 0.009 and remission samples; p < 0.001, respectively). In addition, the frequency of IL-17+ Treg cells among Treg cells was higher during remission than during an attack (uncorrected p = 0.032). Among the lymphocyte subsets, B10 cells alone showed a positive correlation with the intensity of AQP4-ab positivity (ρ [rho] = 0.402 and p = 0.031). It was suggested that the suppressive subsets including B10 and IL-17+ Treg cells might have important roles in controlling disease status in NMOSD. Further functional studies may help to elucidate the immunological role of B10 and IL-17+ Treg cells in NMOSD.
Collapse
|
34
|
Wang Z, Yan Y. Immunopathogenesis in Myasthenia Gravis and Neuromyelitis Optica. Front Immunol 2017; 8:1785. [PMID: 29312313 PMCID: PMC5732908 DOI: 10.3389/fimmu.2017.01785] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/29/2017] [Indexed: 12/13/2022] Open
Abstract
Myasthenia gravis (MG) and neuromyelitis optica (NMO) are autoimmune channelopathies of the peripheral neuromuscular junction (NMJ) and central nervous system (CNS) that are mainly mediated by humoral immunity against the acetylcholine receptor (AChR) and aquaporin-4 (AQP4), respectively. The diseases share some common features, including genetic predispositions, environmental factors, the breakdown of tolerance, the collaboration of T cells and B cells, imbalances in T helper 1 (Th1)/Th2/Th17/regulatory T cells, aberrant cytokine and antibody secretion, and complement system activation. However, some aspects of the immune mechanisms are unique. Both targets (AChR and AQP4) are expressed in the periphery and CNS, but MG mainly affects the NMJ in the periphery outside of CNS, whereas NMO preferentially involves the CNS. Inflammatory cells, including B cells and macrophages, often infiltrate the thymus but not the target—muscle in MG, whereas the infiltration of inflammatory cells, mainly polymorphonuclear leukocytes and macrophages, in NMO, is always observed in the target organ—the spinal cord. A review of the common and discrepant characteristics of these two autoimmune channelopathies may expand our understanding of the pathogenic mechanism of both disorders and assist in the development of proper treatments in the future.
Collapse
Affiliation(s)
- Zhen Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China.,Tianjin Medical University General Hospital, Tianjin Neurological Institute, Tianjin, China
| | - Yaping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
35
|
Murta V, Ferrari C. Peripheral Inflammation and Demyelinating Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:263-285. [PMID: 27714694 DOI: 10.1007/978-3-319-40764-7_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In recent decades, several neurodegenerative diseases have been shown to be exacerbated by systemic inflammatory processes. There is a wide range of literature that demonstrates a clear but complex relationship between the central nervous system (CNS) and the immunological system, both under naïve or pathological conditions. In diseased brains, peripheral inflammation can transform "primed" microglia into an "active" state, which can trigger stronger pathological responses. Demyelinating diseases are a group of neurodegenerative diseases characterized by inflammatory lesions associated with demyelination, which in turn induces axonal damage, neurodegeneration, and progressive loss of function. Among them, the most important are multiple sclerosis (MS) and neuromyelitis optica (NMO). In this review, we will analyze the effect of specific peripheral inflammatory stimuli in the progression of demyelinating diseases and discuss their animal models. In most cases, peripheral immune stimuli are exacerbating.
Collapse
Affiliation(s)
- Verónica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carina Ferrari
- Instituto de Ciencias Básicas y Medicina Experimental, Instituto Universitario del Hospital Italiano, Buenos Aires, Argentina.
| |
Collapse
|
36
|
Han J, Sun L, Wang Z, Fan X, Wang L, Song YY, Zhu J, Jin T. Circulating regulatory B cell subsets in patients with neuromyelitis optica spectrum disorders. Neurol Sci 2017; 38:1205-1212. [PMID: 28389940 DOI: 10.1007/s10072-017-2932-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/23/2017] [Indexed: 12/19/2022]
Abstract
This study analyzed the populations of three different subsets of regulatory B cells (Bregs) in the peripheral blood mononuclear cells (PBMCs) of patients with neuromyelitis optica spectrum disorders (NMOSDs) and explored the relationship between the changes in these subsets of Bregs and the severity of NMOSD. A total of 22 patients with relapsed NMOSDs before treatment were recruited in our study, along with 20 age and gender-matched healthy controls, from May 2015 to March 2016. The percentages and numbers for three different subsets of Bregs including the CD19+CD24hiCD38hi, CD19+CD24hiCD27+, and CD19+CD5+CD1dhi populations were evaluated in parallel by flow cytometry. Afterwards, correlations between the change of three different subsets of Bregs and disease severity were analyzed. We found significantly lower percentages of CD19+CD24hiCD38hi and CD19+CD5+CD1dhi Bregs in NMOSDs patients than in healthy individuals. In contrast, the CD19+CD24hiCD27+ Bregs population was significantly higher in NMOSDs patients than in healthy individuals. However, the three different Bregs subsets showed no significant correlation with expanded disability status scale (EDSS) or annualized relapse rate (ARR). Our findings suggest that the subsets of Bregs may play complex roles in the pathogenesis of NMOSDs and are not correlated with clinical disease severity. Further insights into the potential role of subsets of Bregs could increase our basic knowledge of NMOSDs pathogenesis.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Zhongkun Wang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Xueli Fan
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Lifang Wang
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Yang-Yang Song
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, the First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
37
|
Wang S, Yang T, Wan J, Zhang Y, Fan Y. Elevated C-X-C motif ligand 13 and B-cell-activating factor levels in neuromyelitis optica during remission. Brain Behav 2017; 7:e00648. [PMID: 28413701 PMCID: PMC5390833 DOI: 10.1002/brb3.648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Discovery of specific antibodies against astrocytic water channel aquaporin-4 (AQP4), which is produced by plasma cells, in the serum of neuromyelitis optica (NMO) confirmed the pathogenic role of B cells in NMO. C-X-C motif ligand 13 (CXCL13) and B-cell-activating factor (BAFF) are two crucial factors for antibody production. Relevant studies have focused on the acute phase of NMO. However, CXCL13 and BAFF levels during remission, remain to be elucidated. OBJECTIVE To evaluate serum levels of CXCL13 and BAFF in NMO and multiple sclerosis (MS) patients during remission and explore their correlation with immunosuppressive agents and clinical features in NMO. METHODS Serum CXCL13 and BAFF were measured by enzyme-linked immunosorbent assay (ELISA) in NMO patients, MS patients, and controls. RESULTS Serum CXCL13 levels of NMO patients (n = 24) were significantly higher than those of controls (n = 22) (p = .001), but CXCL13 levels of MS patients (n = 20) and controls (n = 22) did not differ significantly (p = .279). Although the three groups showed no differences in serum BAFF levels, serum BAFF levels of NMO patients without immunosuppressive treatment (n = 8) were significantly elevated compared with those of NMO patients with immunosuppressive therapy (n = 16) (p = .003) and controls (n = 22) (p = .024). In NMO patients, CXCL13 levels were correlated with onset age (p = .026) and duration to the last relapse (p = .003). CONCLUSION During remission, serum CXCL13 and BAFF levels have not decreased to normal in NMO patients, and B-cell-related autoimmune response persists. Immunosuppressive therapy decreased serum BAFF levels, but did not affect CXCL13 expression.
Collapse
Affiliation(s)
- Su Wang
- Department of Traditional Chinese Medicine Beijing Tiantan Hospital Capital Medical University Beijing China.,Department of Oncology Qingdao Hiser Medical Group Qingdao China
| | - Tao Yang
- Department of Traditional Chinese Medicine Beijing Tiantan Hospital Capital Medical University Beijing China
| | - Jianglong Wan
- Department of Traditional Chinese Medicine Beijing Tiantan Hospital Capital Medical University Beijing China.,Department of Traditional Chinese Medicine Miyun Xitiangezhuang Town Community Health Service Center Beijing China
| | - Yongchao Zhang
- Department of Traumatic Orthopedics Qingdao Hiser Medical Group Qingdao China
| | - Yongping Fan
- Department of Traditional Chinese Medicine Beijing Tiantan Hospital Capital Medical University Beijing China
| |
Collapse
|
38
|
Raised cerebrospinal fluid BAFF and APRIL levels in anti- N -methyl- d -aspartate receptor encephalitis: Correlation with clinical outcome. J Neuroimmunol 2017; 305:84-91. [DOI: 10.1016/j.jneuroim.2017.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/29/2016] [Accepted: 01/13/2017] [Indexed: 12/20/2022]
|
39
|
Kalampokis I, Venturi GM, Poe JC, Dvergsten JA, Sleasman JW, Tedder TF. The Regulatory B Cell Compartment Expands Transiently During Childhood and Is Contracted in Children With Autoimmunity. Arthritis Rheumatol 2017; 69:225-238. [PMID: 27429419 PMCID: PMC5195882 DOI: 10.1002/art.39820] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/14/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Regulatory B cells that inhibit immune responses through interleukin-10 (IL-10) secretion (B10 cells) have been characterized in adult subjects with autoimmune disease. The aim of this study was to characterize B10 cells in individuals across the entire age range of normal human development and changes in their frequency and numbers in children with autoimmunity. METHODS The phenotype and numbers of B10 cells in blood were examined in healthy individuals and children with autoimmunity, using flow cytometry. B10 cell function was assessed by measuring the effect of B cell-derived IL-10 on interferon-γ (IFNγ) expression by CD4+ T cells. Serum cytokine levels were measured by enzyme-linked immunosorbent assay. RESULTS The frequency of B10 cells transiently increased during childhood, when up to 30% of B cells were competent to produce IL-10, compared with the low frequencies in healthy newborns (3-4%) and adults (7-9%). The surface phenotype of B10 cells in children revealed age-dependent variability. B10 cells from children were distinct from proinflammatory cytokine-producing B cells and down-regulated IFNγ production by CD4+ T cells in vitro. Compared with age-matched healthy controls, children with autoimmunity had lower numbers and frequencies of B10 cells (decreased by 39% and 48%, respectively), higher IFNγ levels, and lower IL-21 levels in serum. IFNγ inhibited, whereas IL-21 promoted, B cell IL-10 competence in vitro. CONCLUSION B10 cells, a functionally defined cell subset with a variable surface phenotype reflective of overall B cell development, transiently expand during childhood. B10 cell frequencies and numbers were decreased in children with autoimmunity, which may be explained in part by alterations in serum IFNγ and IL-21 that differentially regulate B10 cell development.
Collapse
Affiliation(s)
- Ioannis Kalampokis
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | | | - Jonathan C. Poe
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| | | | - John W. Sleasman
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710
| | - Thomas F. Tedder
- Department of Immunology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
40
|
Chen D, Gallagher S, Monson NL, Herbst R, Wang Y. Inebilizumab, a B Cell-Depleting Anti-CD19 Antibody for the Treatment of Autoimmune Neurological Diseases: Insights from Preclinical Studies. J Clin Med 2016; 5:jcm5120107. [PMID: 27886126 PMCID: PMC5184780 DOI: 10.3390/jcm5120107] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/14/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
Exaggerated or inappropriate responses by B cells are an important feature in many types of autoimmune neurological diseases. The recent success of B-cell depletion in the treatment of multiple sclerosis (MS) has stimulated the development of novel B-cell-targeting therapies with the potential for improved efficacy. CD19 has emerged as a promising target for the depletion of B cells as well as CD19-positive plasmablasts and plasma cells. Inebilizumab (MEDI-551), an anti-CD19 antibody with enhanced antibody-dependent cell-mediated cytotoxicity against B cells, is currently being evaluated in MS and neuromyelitis optica. This review discusses the role of B cells in autoimmune neurological disorders, summarizes the development of inebilizumab, and analyzes the recent results for inebilizumab treatment in an autoimmune encephalitis mouse model. The novel insights obtained from these preclinical studies can potentially guide future investigation of inebilizumab in patients.
Collapse
Affiliation(s)
- Ding Chen
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Sandra Gallagher
- Department of Project Management, MedImmune, Gaithersburg, MD 20878, USA.
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Ronald Herbst
- Department of Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| | - Yue Wang
- Department of Oncology Research, MedImmune, Gaithersburg, MD 20878, USA.
| |
Collapse
|
41
|
Han J, Sun L, Fan X, Wang Z, Cheng Y, Zhu J, Jin T. Role of regulatory b cells in neuroimmunologic disorders. J Neurosci Res 2016; 94:693-701. [PMID: 27112131 PMCID: PMC5074285 DOI: 10.1002/jnr.23749] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 01/02/2023]
Abstract
B lymphocytes augment the immune response by producing antibodies and activating T cells by antigen presentation. Recent studies have highlighted a specific and functionally significant B‐cell subset that could downregulate excessive immune and inflammatory responses through a vast array of inhibitory cytokines, such as interleukin (IL)‐10 and transforming growth factor‐β (TGF‐β). This subset of B cells is generally referred to as regulatory B cells (Bregs). In addition, recent studies have shown that IL‐35‐producing Bregs also play a role in downregulation of immunity. Diverse phenotypes of Bregs have been proposed to underlie human disorders and their animal models. Most studies have focused on the role of different subsets of Bregs and Bregs‐associated molecules such as IL‐10, TGF‐β, and IL‐35 in the pathogenesis of neuroimmunologic disorders. Furthermore, Bregs exert regulatory function mainly through suppressing the differentiation of Th1/Th17 cells and promoting regulatory T‐cell expansion. Reduced presence of Bregs is reportedly associated with progression of several neuroimmunologic disorders. This Review summarizes the current knowledge on the role of Bregs in neuroimmunologic disorders, including multiple sclerosis, neuromyelitis optica, and myasthenia gravis. © 2016 The Authors. Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xueli Fan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Zhongkun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yun Cheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences, and Society, Karolinska Institute, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Alexopoulos H, Biba A, Dalakas MC. Anti-B-Cell Therapies in Autoimmune Neurological Diseases: Rationale and Efficacy Trials. Neurotherapeutics 2016; 13:20-33. [PMID: 26566961 PMCID: PMC4720683 DOI: 10.1007/s13311-015-0402-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antibody-producing cells and, most importantly, as sensors, coordinators, and regulators of the immune response. B cells, among other functions, regulate the T-cell activation process through their participation in antigen presentation and production of cytokines. The availability of monoclonal antibodies or fusion proteins against B-cell surface molecules or B-cell trophic factors bestows a rational approach for treating autoimmune neurological disorders, even when T cells are the main effector cells. This review summarizes basic aspects of B-cell biology, discusses the role(s) of B cells in neurological autoimmunity, and presents anti-B-cell drugs that are either currently on the market or are expected to be available in the near future for treating neurological autoimmune disorders.
Collapse
Affiliation(s)
- Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Angie Biba
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Claes N, Fraussen J, Stinissen P, Hupperts R, Somers V. B Cells Are Multifunctional Players in Multiple Sclerosis Pathogenesis: Insights from Therapeutic Interventions. Front Immunol 2015; 6:642. [PMID: 26734009 PMCID: PMC4685142 DOI: 10.3389/fimmu.2015.00642] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/07/2015] [Indexed: 01/07/2023] Open
Abstract
Multiple sclerosis (MS) is a severe disease of the central nervous system (CNS) characterized by autoimmune inflammation and neurodegeneration. Historically, damage to the CNS was thought to be mediated predominantly by activated pro-inflammatory T cells. B cell involvement in the pathogenesis of MS was solely attributed to autoantibody production. The first clues for the involvement of antibody-independent B cell functions in MS pathology came from positive results in clinical trials of the B cell-depleting treatment rituximab in patients with relapsing-remitting (RR) MS. The survival of antibody-secreting plasma cells and decrease in T cell numbers indicated the importance of other B cell functions in MS such as antigen presentation, costimulation, and cytokine production. Rituximab provided us with an example of how clinical trials can lead to new research opportunities concerning B cell biology. Moreover, analysis of the antibody-independent B cell functions in MS has gained interest since these trials. Limited information is present on the effects of current immunomodulatory therapies on B cell functions, although effects of both first-line (interferon, glatiramer acetate, dimethyl fumarate, and teriflunomide), second-line (fingolimod, natalizumab), and even third-line (monoclonal antibody therapies) treatments on B cell subtype distribution, expression of functional surface markers, and secretion of different cytokines by B cells have been studied to some extent. In this review, we summarize the effects of different MS-related treatments on B cell functions that have been described up to now in order to find new research opportunities and contribute to the understanding of the pathogenesis of MS.
Collapse
Affiliation(s)
- Nele Claes
- Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, School of Life Sciences , Diepenbeek , Belgium
| | - Judith Fraussen
- Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, School of Life Sciences , Diepenbeek , Belgium
| | - Piet Stinissen
- Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, School of Life Sciences , Diepenbeek , Belgium
| | - Raymond Hupperts
- Department of Neuroscience, School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; Department of Neurology, Academic MS Center Limburg, Zuyderland Medisch Centrum, Sittard, Netherlands
| | - Veerle Somers
- Hasselt University, Biomedical Research Institute and Transnationale Universiteit Limburg, School of Life Sciences , Diepenbeek , Belgium
| |
Collapse
|
44
|
Kannel K, Alnek K, Vahter L, Gross-Paju K, Uibo R, Kisand KV. Changes in Blood B Cell-Activating Factor (BAFF) Levels in Multiple Sclerosis: A Sign of Treatment Outcome. PLoS One 2015; 10:e0143393. [PMID: 26600308 PMCID: PMC4658115 DOI: 10.1371/journal.pone.0143393] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 11/04/2015] [Indexed: 01/21/2023] Open
Abstract
Multiple sclerosis (MS) is mediated primarily by autoreactive T cells. However, evidence suggesting the involvement of humoral immunity in brain diseases has increased interest in the role of B cells and their products during MS pathogenesis. The major survival factor for B cells, BAFF has been shown to play a role in several autoimmune conditions. Elevated BAFF levels have been reported in MS animal model and during MS relapse in patients. Moreover, disease-modifying treatments (DMT) reportedly influence blood BAFF levels in MS patients, but the significance of these changes remains unclear. The present study addresses how blood BAFF levels are associated with the clinical course of relapsing-remitting MS and the effectiveness of DMT and short-term steroid treatment. During a prospective longitudinal follow-up of 2.3 years, BAFF was measured in the blood of 170 MS patients in the stable phase and within 186 relapses. BAFF levels were significantly higher in MS patients compared to healthy controls. However, stable MS patients without relapses exhibited significantly higher BAFF levels than relapsing patients. Treatment with interferon-β and immunosuppressants raised BAFF blood levels. Interestingly, a similar effect was not seen in patients treated with glatiramer acetate. Short-term treatment with high doses of intravenous methylprednisolone did not significantly alter plasma BAFF levels in 65% of relapsing-remitting MS patients. BAFF were correlated weakly but significantly with monocyte and basophil counts, but not with other blood cell types (neutrophils, lymphocytes, or eosinophils) or inflammatory biomarkers. To our knowledge, this is the first report demonstrating that higher blood BAFF levels may reflect a more stable and effective MS treatment outcome. These results challenge hypotheses suggesting that elevated blood BAFF levels are associated with more severe disease presentation and could explain the recent failure of pharmaceutical trials targeting BAFF with soluble receptor for MS treatment.
Collapse
Affiliation(s)
- Karin Kannel
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
| | - Kristi Alnek
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
| | - Liina Vahter
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
- Institute of Psychology, Tallinn University, Tallinn, Estonia
| | - Katrin Gross-Paju
- West-Tallinn Central Hospital MS Centre, Tallinn, Estonia
- Institute of Clinical Medicine, Tallinn University of Technology, Tallinn, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
| | - Kalle V. Kisand
- Department of Immunology, Institute of Biomedicine and Translational Medicine, Tartu University, Tartu, Estonia
- * E-mail:
| |
Collapse
|
45
|
Melamed E, Levy M, Waters PJ, Sato DK, Bennett JL, John GR, Hooper DC, Saiz A, Bar-Or A, Kim HJ, Pandit L, Leite MI, Asgari N, Kissani N, Hintzen R, Marignier R, Jarius S, Marcelletti J, Smith TJ, Yeaman MR, Han MH, Aktas O, Apiwattanakul M, Banwell B, Bichuetti D, Broadley S, Cabre P, Chitnis T, De Seze J, Fujihara K, Greenberg B, Hellwig K, Iorio R, Jarius S, Klawiter E, Kleiter I, Lana-Peixoto M, Nakashima, O'Connor K, Palace J, Paul F, Prayoonwiwat N, Ruprecht K, Stuve O, Tedder T, Tenembaum S, Garrahan JP, Aires B, van Herle K, van Pelt D, Villoslada P, Waubant E, Weinshenker B, Wingerchuk D, Würfel J, Zamvil S. Update on biomarkers in neuromyelitis optica. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e134. [PMID: 26236760 PMCID: PMC4516398 DOI: 10.1212/nxi.0000000000000134] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 06/01/2015] [Indexed: 11/15/2022]
Abstract
Neuromyelitis optica (NMO) (and NMO spectrum disorder) is an autoimmune inflammatory disease of the CNS primarily affecting spinal cord and optic nerves. Reliable and sensitive biomarkers for onset, relapse, and progression in NMO are urgently needed because of the heterogeneous clinical presentation, severity of neurologic disability following relapses, and variability of therapeutic response. Detecting aquaporin-4 (AQP4) antibodies (AQP4-IgG or NMO-IgG) in serum supports the diagnosis of seropositive NMO. However, whether AQP4-IgG levels correlate with disease activity, severity, response to therapy, or long-term outcomes is unclear. Moreover, biomarkers for patients with seronegative NMO have yet to be defined and validated. Collaborative international studies hold great promise for establishing and validating biomarkers that are useful in therapeutic trials and clinical management. In this review, we discuss known and potential biomarkers for NMO.
Collapse
Affiliation(s)
- Esther Melamed
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Michael Levy
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Patrick J Waters
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Douglas Kazutoshi Sato
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Jeffrey L Bennett
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Gareth R John
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Douglas C Hooper
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Albert Saiz
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Amit Bar-Or
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Ho Jin Kim
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Lakha Pandit
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Maria Isabel Leite
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Nasrin Asgari
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Najib Kissani
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Rogier Hintzen
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Romain Marignier
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Sven Jarius
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - John Marcelletti
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Terry J Smith
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - Michael R Yeaman
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | - May H Han
- Stanford University (E.M., M.H.H.), Stanford, CA; Johns Hopkins University (M.L.), Baltimore, MD; University of Oxford (P.J.W.), UK; Tohoku University (D.K.S.), Sendai, Japan; University of São Paulo (D.K.S.), Brazil; University of Colorado (J.L.B.), Denver; Mt. Sinai University (G.R.J.), New York, NY; Thomas Jefferson University (D.C.H.), Philadelphia, PA; IDIBAPS (A.S.), Barcelona, Spain; Montreal Neurological Institute and Hospital (A.B.-O.), McGill University, Montreal, Quebec, Canada; Research Institute and Hospital of National Cancer Center (H.J.K.), Goyang, Korea; KS Hegde Medical Academy (L.P.), Nitte University, Mangalore, India; Oxford University Hospital (M.I.L.), Oxford, UK; University of Southern Denmark (N.A.), Odense; Vejle Hospital (N.A.), Denmark; University Hospital (N.K.), Marrakech, Morocco; MS Center (R.H.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Service de Neurologie A (R.M.), Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France; Molecular Neuroimmunology (S.J.), Department of Neurology, University Hospital Heidelberg, Germany; Tandem Labs (J.M.), San Diego, CA; University of Michigan Medical School (T.J.S.), Ann Arbor, MI; and David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Riedhammer C, Weissert R. Antigen Presentation, Autoantigens, and Immune Regulation in Multiple Sclerosis and Other Autoimmune Diseases. Front Immunol 2015; 6:322. [PMID: 26136751 PMCID: PMC4470263 DOI: 10.3389/fimmu.2015.00322] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022] Open
Abstract
Antigen presentation is in the center of the immune system, both in host defense against pathogens, but also when the system is unbalanced and autoimmune diseases like multiple sclerosis (MS) develop. It is not just by chance that a major histocompatibility complex gene is the major genetic susceptibility locus in MS; a feature that MS shares with other autoimmune diseases. The exact etiology of the disease, however, has not been fully understood yet. T cells are regarded as the major players in the disease, but most probably a complex interplay of altered central and peripheral tolerance mechanisms, T-cell and B-cell functions, characteristics of putative autoantigens, and a possible interference of environmental factors like microorganisms are at work. In this review, new data on all these different aspects of antigen presentation and their role in MS will be discussed, probable autoantigens will be summarized, and comparisons to other autoimmune diseases will be drawn.
Collapse
Affiliation(s)
- Christine Riedhammer
- Neuroimmunology, Department of Neurology, University of Regensburg , Regensburg , Germany
| | - Robert Weissert
- Neuroimmunology, Department of Neurology, University of Regensburg , Regensburg , Germany
| |
Collapse
|
47
|
Changes in B and T-cell subsets and NMO-IgG/AQP-4 levels after immunoglobulins and rituximab treatment for an acute attack of neuromyelitis optica. NEUROLOGÍA (ENGLISH EDITION) 2015. [DOI: 10.1016/j.nrleng.2013.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
48
|
Ireland SJ, Monson NL, Davis LS. Seeking balance: Potentiation and inhibition of multiple sclerosis autoimmune responses by IL-6 and IL-10. Cytokine 2015; 73:236-44. [PMID: 25794663 PMCID: PMC4437890 DOI: 10.1016/j.cyto.2015.01.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/12/2015] [Accepted: 01/22/2015] [Indexed: 01/07/2023]
Abstract
The cytokines IL-6 and IL-10 are produced by cells of the adaptive and innate arms of the immune system and they appear to play key roles in genetically diverse autoimmune diseases such as relapsing remitting multiple sclerosis (MS), rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). Whereas previous intense investigations focused on the generation of autoantibodies and their contribution to immune-mediated pathogenesis in these diseases; more recent attention has focused on the roles of cytokines such as IL-6 and IL-10. In response to pathogens, antigen presenting cells (APC), including B cells, produce IL-6 and IL-10 in order to up-or down-regulate immune cell activation and effector responses. Evidence of elevated levels of the proinflammatory cytokine IL-6 has been routinely observed during inflammatory responses and in a number of autoimmune diseases. Our recent studies suggest that MS peripheral blood B cells secrete higher quantities of IL-6 and less IL-10 than B cells from healthy controls. Persistent production of IL-6, in turn, contributes to T cell expansion and the functional hyperactivity of APC such as MS B cells. Altered B cell activity can have a profound impact on resultant T cell effector functions. Enhanced signaling through the IL-6 receptor can effectively inhibit cytolytic activity, induce T cell resistance to IL-10-mediated immunosuppression and increase skewing of autoreactive T cells to a pathogenic Th17 phenotype. Our recent findings and studies by others support a role for the indirect attenuation of B cell responses by Glatiramer acetate (GA) therapy. Our studies suggest that GA therapy temporarily permits homeostatic regulatory mechanisms to be reinstated. Future studies of mechanisms underlying dysregulated B cell cytokine production could lead to the identification of novel targets for improved immunoregulatory therapies for autoimmune diseases.
Collapse
Affiliation(s)
- Sara J Ireland
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| | - Nancy L Monson
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| | - Laurie S Davis
- Rheumatic Diseases Division, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, United States.
| |
Collapse
|
49
|
Bennett JL, O'Connor KC, Bar-Or A, Zamvil SS, Hemmer B, Tedder TF, von Büdingen HC, Stuve O, Yeaman MR, Smith TJ, Stadelmann C. B lymphocytes in neuromyelitis optica. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2015; 2:e104. [PMID: 25977932 PMCID: PMC4426682 DOI: 10.1212/nxi.0000000000000104] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/16/2015] [Indexed: 12/21/2022]
Abstract
Neuromyelitis optica (NMO) is an inflammatory autoimmune disorder of the CNS that predominantly affects the spinal cord and optic nerves. A majority (approximately 75%) of patients with NMO are seropositive for autoantibodies against the astrocyte water channel aquaporin-4 (AQP4). These autoantibodies are predominantly IgG1, and considerable evidence supports their pathogenicity, presumably by binding to AQP4 on CNS astrocytes, resulting in astrocyte injury and inflammation. Convergent clinical and laboratory-based investigations have indicated that B cells play a fundamental role in NMO immunopathology. Multiple mechanisms have been hypothesized: AQP4 autoantibody production, enhanced proinflammatory B cell and plasmablast activity, aberrant B cell tolerance checkpoints, diminished B cell regulatory function, and loss of B cell anergy. Accordingly, many current off-label therapies for NMO deplete B cells or modulate their activity. Understanding the role and mechanisms whereby B cells contribute to initiation, maintenance, and propagation of disease activity is important to advancing our understanding of NMO pathogenesis and developing effective disease-specific therapies.
Collapse
Affiliation(s)
- Jeffrey L Bennett
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Kevin C O'Connor
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Amit Bar-Or
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Scott S Zamvil
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Bernhard Hemmer
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Thomas F Tedder
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - H-Christian von Büdingen
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Olaf Stuve
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Michael R Yeaman
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Terry J Smith
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| | - Christine Stadelmann
- Departments of Neurology and Ophthalmology and Neuroscience Program (J.L.B.), University of Colorado, Denver; Department of Neurology (K.C.O.), Yale University School of Medicine, New Haven, CT; Neuroimmunology Unit (A.B.-O.), Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology (S.S.Z., H.-C.v.B.), UCSF School of Medicine, San Francisco, CA; Department of Neurology (B.H.), Technische Universität München, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Immunology (T.F.T.), Duke University Medical Center, Durham, NC; Departments of Neurology and Neurotherapeutics (O.S.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Medicine (M.R.Y.), Divisions of Molecular Medicine and Infectious Diseases, University of California, Los Angeles; Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA; Departments of Ophthalmology and Visual Sciences and Internal Medicine (T.J.S.), University of Michigan Medical School, Ann Arbor; and Institute of Neuropathology (C.S.), University Medical Center, Göttingen, Germany
| |
Collapse
|
50
|
Quan C, ZhangBao J, Lu J, Zhao C, Cai T, Wang B, Yu H, Qiao J, Lu C. The immune balance between memory and regulatory B cells in NMO and the changes of the balance after methylprednisolone or rituximab therapy. J Neuroimmunol 2015; 282:45-53. [PMID: 25903728 DOI: 10.1016/j.jneuroim.2015.03.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 03/03/2015] [Accepted: 03/20/2015] [Indexed: 10/23/2022]
Abstract
We aim to explore the impacts of high dose methylprednisolone therapy (HDMT) and rituximab on circulating B cells in NMO patients. Twenty-two NMO patients in the acute relapse phase were treated with HDMT and 9 patients in the remission stage were treated with rituximab. The frequencies of circulating CD19(+)CD27(+) memory B cell (Bmem), CD19(+)CD24(high)CD38(high) regulatory B cell (Breg) and the B cell production of interleukin (IL)-10 and interferon (IFN)-γ were monitored by flow cytometry before and after the treatment. The frequencies of circulating Bregs and the B cell IL-10 production were significantly lower in NMO patients before HDMT compared to healthy controls. Two weeks' HDMT further reduced the frequencies of Bregs while increased the frequencies of Bmems, which steered the numerical balance between Bmem and Breg in favor of Bmem. Meanwhile, HDMT significantly inhibited the B cell IFN-γ expression. Rituximab exerted its effect through B cell elimination and subsequent B cell repopulation which was characterized by the predominance of Bregs, restored the numerical balance between Breg and Bmem back to an advantageous "Breg>Bmem" status. Therefore, HDMT and rituximab had basically different impacts on B cells in NMO.
Collapse
Affiliation(s)
- Chao Quan
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jingzi ZhangBao
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China; Department of Neurology, Jing'an District Centre Hospital of Shanghai, China
| | - Tongjia Cai
- Department of Neurology, Jing'an District Centre Hospital of Shanghai, China
| | - Bei Wang
- Department of Neurology, Jing'an District Centre Hospital of Shanghai, China
| | - Hai Yu
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Jian Qiao
- Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China
| | - Chuanzhen Lu
- Department and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, China.
| |
Collapse
|