1
|
Xu L, Xu H, Tang C. Aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders: progress of experimental models based on disease pathogenesis. Neural Regen Res 2025; 20:354-365. [PMID: 38819039 PMCID: PMC11317952 DOI: 10.4103/nrr.nrr-d-23-01325] [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: 08/08/2023] [Revised: 11/18/2023] [Accepted: 12/19/2023] [Indexed: 06/01/2024] Open
Abstract
Neuromyelitis optica spectrum disorders are neuroinflammatory demyelinating disorders that lead to permanent visual loss and motor dysfunction. To date, no effective treatment exists as the exact causative mechanism remains unknown. Therefore, experimental models of neuromyelitis optica spectrum disorders are essential for exploring its pathogenesis and in screening for therapeutic targets. Since most patients with neuromyelitis optica spectrum disorders are seropositive for IgG autoantibodies against aquaporin-4, which is highly expressed on the membrane of astrocyte endfeet, most current experimental models are based on aquaporin-4-IgG that initially targets astrocytes. These experimental models have successfully simulated many pathological features of neuromyelitis optica spectrum disorders, such as aquaporin-4 loss, astrocytopathy, granulocyte and macrophage infiltration, complement activation, demyelination, and neuronal loss; however, they do not fully capture the pathological process of human neuromyelitis optica spectrum disorders. In this review, we summarize the currently known pathogenic mechanisms and the development of associated experimental models in vitro, ex vivo, and in vivo for neuromyelitis optica spectrum disorders, suggest potential pathogenic mechanisms for further investigation, and provide guidance on experimental model choices. In addition, this review summarizes the latest information on pathologies and therapies for neuromyelitis optica spectrum disorders based on experimental models of aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders, offering further therapeutic targets and a theoretical basis for clinical trials.
Collapse
Affiliation(s)
- Li Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
Krett JD, Filippatou AG, Barreras P, Pardo CA, Gelber AC, Sotirchos ES. "Lupus Myelitis" Revisited: A Retrospective Single-Center Study of Myelitis Associated With Rheumatologic Disease. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200329. [PMID: 39442039 DOI: 10.1212/nxi.0000000000200329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND OBJECTIVES Previous reports of patients with myelitis associated with rheumatologic disease may have had unrecognized aquaporin-4 (AQP4)-IgG seropositive neuromyelitis optica spectrum disorder (NMOSD) or myelin oligodendrocyte glycoprotein (MOG)-IgG-associated disease (MOGAD). We clinicoradiologically and serologically characterized patients with myelitis associated with rheumatologic disease evaluated in the era of availability of MOG-IgG and more sensitive AQP4-IgG cell-based assays. METHODS A retrospective cohort (2018-2023) at Johns Hopkins Medicine with diagnoses of myelopathy and rheumatologic comorbidity was identified by electronic medical record (EMR) query. All patients with myelitis unrelated to typical multiple sclerosis (MS) were included and analyzed by chart review. RESULTS Of 238 patients identified by EMR query, 197 were excluded (148 not meeting prespecified inclusion criteria, 49 had typical MS), resulting in 41 patients for review. The mean age at myelitis onset was 44 ± 15 years; 39 (95%) were female. Rheumatologic diagnoses included 17 (41.5%) with systemic lupus erythematosus (SLE), 10 (24.3%) Sjögren syndrome (SS), 6 (15%) undifferentiated connective tissue disease (UCTD), 5 (12%) combinations of SLE/SS/UCTD with antiphospholipid antibody syndrome, 1 (2.4%) rheumatoid arthritis, 1 (2.4%) psoriatic arthritis, and 1 (2.4%) Behçet disease. 20 patients (49%) were diagnosed with AQP4-IgG seropositive NMOSD, 3 (7%) with MOGAD, and 18 (44%) had "double-seronegative" myelitis. Of these 18, 3 were diagnosed with AQP4-IgG seronegative NMOSD, 1 neuro-Behçet disease, and 14 other (unclassifiable) myelitis. Excluding 1 patient with neuro-Behçet disease, 18 (90%) of 20 AQP4-IgG seropositive patients had longitudinally extensive cord lesions compared with 5 (29%; p < 0.001) of 17 "double-seronegative" patients and 2 (67%) of 3 with MOGAD. "Double-seronegative" patients more commonly had CSF-restricted oligoclonal bands. Functional outcomes did not differ by diagnosis, and most patients received acute immunotherapy at the time of initial myelitis diagnosis with at least partial recovery over a median follow-up of 38 (interquartile range: 9-74) months. DISCUSSION Approximately half of our rheumatologic disease cohort with myelitis unrelated to MS had AQP4-IgG seropositive NMOSD while MOGAD accounted for a small but clinically relevant proportion of patients. Further research is needed to characterize myelitis etiology in patients who are seronegative for both AQP4-IgG and MOG-IgG.
Collapse
Affiliation(s)
- Jonathan D Krett
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Angeliki G Filippatou
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paula Barreras
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carlos A Pardo
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allan C Gelber
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elias S Sotirchos
- From the Division of Neuroimmunology and Neurological Infections (J.D.K., A.G.F., P.B., C.A.P., E.S.S.), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD; Multiple Sclerosis and Neuroimmunology Center (P.B.), Department of Neurology and Neurosurgery, Cedars-Sinai Medical Center, University of California, Los Angeles; and Division of Rheumatology (A.C.G.), Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
3
|
Huang Y, Wang T, Wang F, Wu Y, Ai J, Zhang Y, Shao M, Fang L. Scientific issues with rodent models of neuromyelitis optic spectrum disorders. Front Immunol 2024; 15:1423107. [PMID: 39628487 PMCID: PMC11611858 DOI: 10.3389/fimmu.2024.1423107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/21/2024] [Indexed: 12/06/2024] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) is a rare autoimmune disorder that causes severe inflammation in the central nervous system (CNS), primarily affecting the optic nerves, spinal cord, and brainstem. Aquaporin-4 immunoglobulin G antibodies (AQP4-IgG) are a diagnostic marker of the disease and play a significant role in its pathogenesis, though the exact mechanism is not yet fully understood. To develop rodent models that best simulate the in vivo pathological and physiological processes of NMOSD, researchers have been continuously exploring how to establish the ideal model. In this process, two key issues arise: 1) how the AQP4 antibody crosses the blood-brain barrier, and 2) the source of the AQP4 antibody. These two factors are critical for the successful development of rodent models of NMOSD. This paper reviews the current state of research on these two aspects.
Collapse
Affiliation(s)
- Yusen Huang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tianwei Wang
- Department of Radiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Fangruyue Wang
- The Third Bethune Hospital of Jilin University, Changchun, China
| | - Yujing Wu
- The Third Bethune Hospital of Jilin University, Changchun, China
| | - Jia Ai
- The Third Bethune Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- The Third Bethune Hospital of Jilin University, Changchun, China
| | - Meiyan Shao
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Le Fang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Shimizu F, Nakamori M. Blood-Brain Barrier Disruption in Neuroimmunological Disease. Int J Mol Sci 2024; 25:10625. [PMID: 39408955 PMCID: PMC11476930 DOI: 10.3390/ijms251910625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
The blood-brain barrier (BBB) acts as a structural and functional barrier for brain homeostasis. This review highlights the pathological contribution of BBB dysfunction to neuroimmunological diseases, including multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), autoimmune encephalitis (AE), and paraneoplastic neurological syndrome (PNS). The transmigration of massive lymphocytes across the BBB caused by the activation of cell adhesion molecules is involved in the early phase of MS, and dysfunction of the cortical BBB is associated with the atrophy of gray matter in the late phase of MS. At the onset of NMOSD, increased permeability of the BBB causes the entry of circulating AQP4 autoantibodies into the central nervous system (CNS). Recent reports have shown the importance of glucose-regulated protein (GRP) autoantibodies as BBB-reactive autoantibodies in NMOSD, which induce antibody-mediated BBB dysfunction. BBB breakdown has also been observed in MOGAD, NPSLE, and AE with anti-NMDAR antibodies. Our recent report demonstrated the presence of GRP78 autoantibodies in patients with MOGAD and the molecular mechanism responsible for GRP78 autoantibody-mediated BBB impairment. Disruption of the BBB may explain the symptoms in the brain and cerebellum in the development of PNS, as it induces the entry of pathogenic autoantibodies or lymphocytes into the CNS through autoimmunity against tumors in the periphery. GRP78 autoantibodies were detected in paraneoplastic cerebellar degeneration and Lambert-Eaton myasthenic syndrome, and they were associated with cerebellar ataxia with anti-P/Q type voltage-gated calcium channel antibodies. This review reports that therapies affecting the BBB that are currently available for disease-modifying therapies for neuroimmunological diseases have the potential to prevent BBB damage.
Collapse
Affiliation(s)
- Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan;
| | | |
Collapse
|
5
|
Kawakami N, Wekerle H. Life history of a brain autoreactive T cell: From thymus through intestine to blood-brain barrier and brain lesion. Neurotherapeutics 2024; 21:e00442. [PMID: 39237437 PMCID: PMC11585894 DOI: 10.1016/j.neurot.2024.e00442] [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: 05/06/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024] Open
Abstract
Brain antigen-specific autoreactive T cells seem to play a key role in inducing inflammation in the central nervous system (CNS), a characteristic feature of human multiple sclerosis (MS). These T cells are generated within the thymus, where they escape negative selection and become integrated into the peripheral immune repertoire of immune cells. Typically, these autoreactive T cells rest in the periphery without attacking the CNS. When autoimmune T cells enter gut-associated lymphatic tissue (GALT), they may be stimulated by the microbiota and its metabolites. After activation, the cells migrate into the CNS through the blood‒brain barrier, become reactivated upon interacting with local antigen-presenting cells, and induce inflammatory lesions within the brain parenchyma. This review describes how microbiota influence autoreactive T cells during their life, starting in the thymus, migrating through the periphery and inducing inflammation in their target organ, the CNS.
Collapse
Affiliation(s)
- Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany.
| | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital, LMU Munich and Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Germany; Emeritus Group Neuroimmunology, Max Planck Institute of Biological Intelligence, Germany.
| |
Collapse
|
6
|
Fang T, Wu W, He X, Liang Y, Lin Q, Dai K, Wang S, Peng F, Jiang Y. Clinical characteristics of overlapping syndrome in patients with GFAP-IgG and MOG-IgG: a case series of 8 patients and literature review. J Neurol 2024; 271:6811-6821. [PMID: 39190107 DOI: 10.1007/s00415-024-12633-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
OBJECTIVE The overlapping syndrome of anti-GFAP and anti-MOG antibodies is extremely rare. This retrospective study reports 8 adult cases of the GFAP-MOG overlapping syndrome. METHODS We reviewed the clinical characteristics of 8 adult patients with the GFAP-MOG overlapping syndrome from Jan 2019 and Sep 2023 at the Third Affiliated Hospital, Sun Yat-sen University. Moreover, we searched the literature and included all case reports with this overlapping syndrome since 2018 on PubMed. RESULTS The predominant clinical syndrome was meningoencephalomyelitis (5/8), followed by meningoencephalitis (2/8), and myelitis (1/8). Five patients had a flu-like prodromal symptom or diarrhea. No neoplasms were found in these patients. Regarding brain MRI, T2-weighted/fluid-attenuated inversion recovery hyperintensities were in 7 patients and leptomeningeal enhancement was in 4 patients. However, only one patient had periventricular radial linear enhancement. Besides, two patients had large space-occupying lesions. For spinal MRI, T2-hyperintensities were observed in 4 patients, in which 3 patients had longitudinally extensive lesions. All patients were treated with immunotherapy, the median follow-up period was 18 months (range, 3-36 months). Three patients presented relapses during the follow-up, but all cases recovered to mRS scores ≤ 2 at last follow-up. In addition, we also reviewed 14 cases (including 7 adults and 7 children) with this overlapping syndrome by literature review. CONCLUSION Our findings provide data to understand the clinical features and prognosis of the GFAP-MOG overlapping syndrome. Recognizing this overlapping syndrome will expand our knowledge, allowing for better management of these patients.
Collapse
Affiliation(s)
- Ting Fang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600# Tianhe Road, Guangzhou, 510630, Guangdong Province, China
- Department of Neurology, Shantou Central Hospital, 114# Waima Road, Shantou, 515031, Guangdong Province, China
| | - Weijuan Wu
- Department of Neurology, Sanshui District People's Hospital, Sanshui, Foshan, 528100, Guangdong Province, China
| | - Xinjie He
- Department of Cardiology, Shantou Central Hospital, 114# Waima Road, Shantou, 515031, Guangdong Province, China
| | - Yuting Liang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Yuedong Hospital, Meizhou, 514799, Guangdong Province, China
| | - Qi Lin
- Department of Neurology, Shantou Central Hospital, 114# Waima Road, Shantou, 515031, Guangdong Province, China
| | - Kai Dai
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600# Tianhe Road, Guangzhou, 510630, Guangdong Province, China
| | - Siguang Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600# Tianhe Road, Guangzhou, 510630, Guangdong Province, China
| | - Fuhua Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600# Tianhe Road, Guangzhou, 510630, Guangdong Province, China.
| | - Ying Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, 600# Tianhe Road, Guangzhou, 510630, Guangdong Province, China.
| |
Collapse
|
7
|
Bouwman HB, Guchelaar HJ. The efficacy and safety of eculizumab in patients and the role of C5 polymorphisms. Drug Discov Today 2024; 29:104134. [PMID: 39111540 DOI: 10.1016/j.drudis.2024.104134] [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: 03/14/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/13/2024]
Abstract
Eculizumab is an orphan drug with indications for extremely rare autoimmune disorders. It is primarily prescribed for use in patients with paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome; but is also highly effective in the treatment of myasthenia gravis, among others. By binding to the C5 protein in the complement system, eculizumab effectively inhibits cellular hemolysis and autoimmune reactions. Despite this effective treatment, some patients reported no improvement in symptoms. Genetic sequencing revealed three distinct C5 mutations in the non-responders and these polymorphisms appeared to be most prevalent among Japanese, Korean and African populations. Here, we present an overview of the current and potential future applications of eculizumab, as well as the disadvantages of eculizumab treatment in patients with C5 polymorphisms.
Collapse
Affiliation(s)
| | - Henk-Jan Guchelaar
- Clinical Pharmacy and Toxicology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| |
Collapse
|
8
|
Rotstein DL, Freedman MS, Konig A, Lee L, Luo J, Maxwell C, Morrow SA, Tremlett H, Vyas MV, Marrie RA. Investigation of health care use and a possible prodrome before the first attack in NMOSD and MOGAD. Mult Scler 2024; 30:1331-1340. [PMID: 39234853 PMCID: PMC11457589 DOI: 10.1177/13524585241272939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Prodromal phases are well recognized in many inflammatory and neurodegenerative diseases, including multiple sclerosis. We evaluated the possibility of a prodrome in aquaporin-4 antibody positive (AQP4+) neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody disease (MOGAD) using health administrative data. METHODS We investigated individuals with AQP4 + NMOSD and MOGAD, confirmed by medical chart review, in Ontario, Canada. Each NMOSD and MOGAD participant was matched 1:5 to general population controls by sex, birth year, immigrant status, and region. Total outpatient visits and hospitalizations were compared in the 5 years preceding the incident attack in multivariable negative binomial models. RESULTS We identified 96 people with AQP4 + NMOSD, matched to 479 controls, and 61 people with MOGAD, matched to 303 controls. In the 5 years preceding the incident attack, health care use was elevated for outpatient visits and hospitalizations for the NMOSD cohort (adjusted rate ratio (aRR): 1.47; 95% confidence interval (CI): 1.25-1.73; aRR: 1.67; 95% CI: 1.19-2.36, respectively) but not for MOGAD. Rate ratios steadily increased in NMOSD for outpatient visits in the 2 years preceding the incident attack. CONCLUSION Our findings support a prodromal phase preceding clinical onset of AQP4 + NMOSD. Earlier recognition and management of NMOSD patients may be possible.
Collapse
Affiliation(s)
- Dalia L Rotstein
- St. Michael’s Hospital, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | | | - Liesly Lee
- Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Jin Luo
- Institute for Clinical Evaluative Sciences, Toronto, ON, Canada
| | - Colleen Maxwell
- Institute for Clinical Evaluative Sciences, Toronto, ON, Canada; School of Pharmacy, University of Waterloo, Waterloo, ON, Canada
| | - Sarah A Morrow
- Western University, London, ON, Canada; University of Calgary, Calgary, AB, Canada
| | - Helen Tremlett
- Division of Neurology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Manav V Vyas
- St. Michael’s Hospital, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruth Ann Marrie
- Departments of Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
9
|
Mireles-Ramírez MA, Pacheco-Moises FP, González-Usigli HA, Sánchez-Rosales NA, Hernández-Preciado MR, Delgado-Lara DLC, Hernández-Cruz JJ, Ortiz GG. Neuromyelitis optica spectrum disorder: pathophysiological approach. Int J Neurosci 2024; 134:826-838. [PMID: 36453541 DOI: 10.1080/00207454.2022.2153046] [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: 03/12/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Aim: To review the main pathological findings of Neuromyelitis Optica Spectrum Disorder (NMOSD) associated with the presence of autoantibodies to aquaporin-4 (AQP4) as well as the mechanisms of astrocyte dysfunction and demyelination. Methods: An comprehensive search of the literature in the field was carried out using the database of The National Center for Biotechnology Information from . Systematic searches were performed until July 2022. Results: NMOSD is an inflammatory and demyelinating disease of the central nervous system mainly in the areas of the optic nerves and spinal cord, thus explaining mostly the clinical findings. Other areas affected in NMOSD are the brainstem, hypothalamus, and periventricular regions. Relapses in NMOSD are generally severe and patients only partially recover. NMOSD includes clinical conditions where autoantibodies to aquaporin-4 (AQP4-IgG) of astrocytes are detected as well as similar clinical conditions where such antibodies are not detected. AQP4 are channel-forming integral membrane proteins of which AQ4 isoforms are able to aggregate in supramolecular assemblies termed orthogonal arrays of particles (OAP) and are essential in the regulation of water homeostasis and the adequate modulation of neuronal activity and circuitry. AQP4 assembly in orthogonal arrays of particles is essential for AQP4-IgG pathogenicity since AQP4 autoantibodies bind to OAPs with higher affinity than for AQP4 tetramers. NMOSD has a complex background with prominent roles for genes encoding cytokines and cytokine receptors. AQP4 autoantibodies activate the complement-mediated inflammatory demyelination and the ensuing damage to AQP4 water channels, leading to water influx, necrosis and axonal loss. Conclusions: NMOSD as an astrocytopathy is a nosological entity different from multiple sclerosis with its own serological marker: immunoglobulin G-type autoantibodies against the AQP4 protein which elicits a complement-dependent cytotoxicity and neuroinflammation. Some patients with typical manifestations of NMSOD are AQP4 seronegative and myelin oligodendrocyte glycoprotein positive. Thus, the detection of autoantibodies against AQP4 or other autoantibodies is crucial for the correct treatment of the disease and immunosuppressant therapy is the first choice.
Collapse
Affiliation(s)
- Mario A Mireles-Ramírez
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Fermín P Pacheco-Moises
- Department of Chemistry, University Center of Exact Sciences and Engineering; University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Héctor A González-Usigli
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Nayeli A Sánchez-Rosales
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Martha R Hernández-Preciado
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | | | - José J Hernández-Cruz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Genaro Gabriel Ortiz
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
10
|
Namatame C, Abe Y, Miyasaka Y, Takai Y, Matsumoto Y, Takahashi T, Mashimo T, Misu T, Fujihara K, Yasui M, Aoki M. Humanized-Aquaporin-4-Expressing Rat Created by Gene-Editing Technology and Its Use to Clarify the Pathology of Neuromyelitis Optica Spectrum Disorder. Int J Mol Sci 2024; 25:8169. [PMID: 39125739 PMCID: PMC11311328 DOI: 10.3390/ijms25158169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Conventional rodent neuromyelitis optica spectrum disorder (NMOSD) models using patient-derived immunoglobulin G (IgG) are potentially affected by the differences between the human and rodent aquaporin-4 (AQP4) extracellular domains (ECDs). We hypothesized that the humanization of AQP4 ECDs would make the rodent model lesions closer to human NMOSD pathology. Humanized-AQP4-expressing (hAQP4) rats were generated using genome-editing technology, and the human AQP4-specific monoclonal antibody (mAb) or six patient-derived IgGs were introduced intraperitoneally into hAQP4 rats and wild-type Lewis (WT) rats after immunization with myelin basic protein and complete Freund's adjuvant. Human AQP4-specific mAb induced astrocyte loss lesions specifically in hAQP4 rats. The patient-derived IgGs also induced NMOSD-like tissue-destructive lesions with AQP4 loss, demyelination, axonal swelling, complement deposition, and marked neutrophil and macrophage/microglia infiltration in hAQP4 rats; however, the difference in AQP4 loss lesion size and infiltrating cells was not significant between hAQP4 and WT rats. The patient-derived IgGs bound to both human and rat AQP4 M23, suggesting their binding to the shared region of human and rat AQP4 ECDs. Anti-AQP4 titers positively correlated with AQP4 loss lesion size and neutrophil and macrophage/microglia infiltration. Considering that patient-derived IgGs vary in binding sites and affinities and some of them may not bind to rodent AQP4, our hAQP4 rat is expected to reproduce NMOSD-like pathology more accurately than WT rats.
Collapse
Affiliation(s)
- Chihiro Namatame
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yoshiki Miyasaka
- Laboratory of Reproductive Engineering, Institute of Experimental Animal Sciences, Osaka University Medical School, Suita 565-0871, Japan
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Yuki Matsumoto
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Toshiyuki Takahashi
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
- Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa 992-1202, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Kazuo Fujihara
- Department of Multiple Sclerosis & Therapeutics, Fukushima Medical University, Fukushima 960-1295, Japan
- Multiple Sclerosis & Neuromyelitis Optica Center, Southern Tohoku Research Institute for Neuroscience, Koriyama 963-8563, Japan
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
11
|
Katsu M, Sekine-Tanaka M, Tanaka M, Horai Y, Akatsuka A, Suga M, Kiyohara K, Fujita T, Sasaki A, Yamashita T. Inhibition of repulsive guidance molecule-a ameliorates compromised blood-spinal cord barrier integrity associated with neuromyelitis optica in rats. J Neuroimmunol 2024; 388:578297. [PMID: 38306928 DOI: 10.1016/j.jneuroim.2024.578297] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
The influx of pathogenic aquaporin-4 antibodies (AQP4-Abs) across the blood-spinal cord barrier (BSCB) is crucial for the development and exacerbation of neuromyelitis optica (NMO). We examined whether prophylactic intravenous administration of anti-repulsive guidance molecule-a antibodies (RGMa-Abs) has disease-modifying effects on BSCB dysfunction using an NMO model elicited by peripheral administration of AQP4-Abs to rats. RGMa-Ab treatment attenuated the acute exacerbation of perivascular astrocytopathy in the spinal cord and clinical symptoms, which were highly correlated with neurofilament light chain levels in both the cerebrospinal fluid (CSF) and serum. Additionally, RGMa-Ab treatment suppressed the expression of proinflammatory cytokines/chemokines and the infiltration of inflammatory cells into the spinal cord. CSF analysis of NMO rats revealed that RGMa-Ab treatment improved the CSF/serum albumin ratio and suppressed AQP4-Abs influx. RGMa inhibition using RGMa-Abs is suggested as a potential therapeutic option for BSCB dysfunction associated with NMO.
Collapse
Affiliation(s)
- Masataka Katsu
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Misuzu Sekine-Tanaka
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Masaharu Tanaka
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Yasushi Horai
- Research Unit/Frontier Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Shonan Health Innovation Park, 2-26-1, Muraoka-Higashi, Fujisawa-shi, Kanagawa 251-8555, Japan.
| | - Airi Akatsuka
- Research Unit/Frontier Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Shonan Health Innovation Park, 2-26-1, Muraoka-Higashi, Fujisawa-shi, Kanagawa 251-8555, Japan.
| | - Misao Suga
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Kazuhiro Kiyohara
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Takuya Fujita
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Atsushi Sasaki
- Research Unit/Neuroscience Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan.
| | - Toshihide Yamashita
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan; Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
12
|
Wang M, Dehlinger A, Zapata CF, Golan M, Gallaccio G, Sander LE, Schlickeiser S, Kunkel D, Schmitz-Hübsch T, Sawitzki B, Karni A, Braun J, Loyal L, Thiel A, Bellmann-Strobl J, Paul F, Meyer-Arndt L, Böttcher C. Associations of myeloid cells with cellular and humoral responses following vaccinations in patients with neuroimmunological diseases. Nat Commun 2023; 14:7728. [PMID: 38007484 PMCID: PMC10676398 DOI: 10.1038/s41467-023-43553-z] [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: 05/17/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023] Open
Abstract
Disease-modifying therapies (DMTs) are widely used in neuroimmunological diseases such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). Although these treatments are known to predispose patients to infections and affect their responses to vaccination, little is known about the impact of DMTs on the myeloid cell compartment. In this study, we use mass cytometry to examine DMT-associated changes in the innate immune system in untreated and treated patients with MS (n = 39) or NMOSD (n = 23). We also investigated the association between changes in myeloid cell phenotypes and longitudinal responsiveness to homologous primary, secondary, and tertiary SARS-CoV-2 mRNA vaccinations. Multiple DMT-associated myeloid cell clusters, in particular CD64+HLADRlow granulocytes, showed significant correlations with B and T cell responses induced by vaccination. Our findings suggest the potential role of myeloid cells in cellular and humoral responses following vaccination in DMT-treated patients with neuroimmunological diseases.
Collapse
Affiliation(s)
- Meng Wang
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Adeline Dehlinger
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Camila Fernández Zapata
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maya Golan
- Neuroimmunology and Multiple Sclerosis Unit and Laboratory, Sourasky Medical Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Gerardina Gallaccio
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Leif E Sander
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stephan Schlickeiser
- Institute of Medical Immunology, BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, and Berlin Institute of Health Berlin, Berlin, Germany
| | - Desiree Kunkel
- Flow&MassCytometry Core Facility, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Birgit Sawitzki
- Translational Immunology, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arnon Karni
- Neuroimmunology and Multiple Sclerosis Unit and Laboratory, Sourasky Medical Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine and Sagol School of Neuroscience Tel Aviv University, Tel Aviv, Israel
| | - Julian Braun
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Lucie Loyal
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Andreas Thiel
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Immunomics-Regenerative Immunology and Aging, Berlin, Germany
| | - Judith Bellmann-Strobl
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lil Meyer-Arndt
- Neuroscience Clinical Research Center, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Si-M / "Der Simulierte Mensch" a science framework of Technische Universität Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
13
|
Remlinger J, Bagnoud M, Meli I, Massy M, Linington C, Chan A, Bennett JL, Hoepner R, Enzmann V, Salmen A. Modelling MOG antibody-associated disorder and neuromyelitis optica spectrum disorder in animal models: Spinal cord manifestations. Mult Scler Relat Disord 2023; 78:104892. [PMID: 37499337 DOI: 10.1016/j.msard.2023.104892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/18/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
Antibodies to myelin oligodendrocyte glycoprotein (MOG-IgG) or aquaporin 4 (AQP4-IgG) are associated with CNS inflammatory disorders. We directly compared MOG35-55-induced experimental autoimmune encephalomyelitis exacerbated by MOG- and AQP4-IgG (versus isotype IgG, Iso-IgG). Disease severity was highest after MOG-IgG application. MOG- and AQP4-IgG administration increased disease incidence compared to Iso-IgG. Inflammatory lesions appeared earlier and with distinct localizations after AQP4-IgG administration. AQP4 intensity was more reduced after AQP4- than MOG-IgG administration at acute disease phase. The described models are suitable for comparative analyses of pathological features associated with MOG- and AQP4-IgG and the investigation of therapeutic interventions.
Collapse
Affiliation(s)
- Jana Remlinger
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Maud Bagnoud
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Ivo Meli
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Marine Massy
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States of America
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland.
| |
Collapse
|
14
|
Wolf HN, Ehinger V, Guempelein L, Banerjee P, Kuempfel T, Havla J, Pauly D. NMOSD IgG Impact Retinal Cells in Murine Retinal Explants. Curr Issues Mol Biol 2023; 45:7319-7335. [PMID: 37754247 PMCID: PMC10529972 DOI: 10.3390/cimb45090463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/28/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are chronic inflammatory diseases of the central nervous system, characterized by autoantibodies against aquaporin-4. The symptoms primarily involve severe optic neuritis and longitudinally extensive transverse myelitis. Although the disease progression is typically relapse-dependent, recent studies revealed retinal neuroaxonal degeneration unrelated to relapse activity, potentially due to anti-aquaporin-4-positive antibodies interacting with retinal glial cells such as Müller cells. In this exploratory study, we analysed the response of mouse retinal explants to NMOSD immunoglobulins (IgG). Mouse retinal explants were treated with purified IgG from patient or control sera for one and three days. We characterized tissue response patterns through morphological changes, chemokine secretion, and complement expression. Mouse retinal explants exhibited a basic proinflammatory response ex vivo, modified by IgG addition. NMOSD IgG, unlike control IgG, increased gliosis and decreased chemokine release (CCL2, CCL3, CCL4, and CXCL-10). Complement component expression by retinal cells remained unaltered by either IgG fraction. We conclude that human NMOSD IgG can possibly bind in the mouse retina, altering the local cellular environment. This intraretinal stress may contribute to retinal degeneration independent of relapse activity in NMOSD, suggesting a primary retinopathy.
Collapse
Affiliation(s)
- Hannah Nora Wolf
- Department of Experimental Ophthalmology, University Marburg, 35037 Marburg, Germany
| | - Veronika Ehinger
- Department of Experimental Ophthalmology, University Marburg, 35037 Marburg, Germany
| | - Larissa Guempelein
- Department of Experimental Ophthalmology, University Marburg, 35037 Marburg, Germany
| | - Pratiti Banerjee
- Department of Experimental Ophthalmology, University Marburg, 35037 Marburg, Germany
| | - Tania Kuempfel
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Diana Pauly
- Department of Experimental Ophthalmology, University Marburg, 35037 Marburg, Germany
| |
Collapse
|
15
|
Boldrini VO, Brito MR, Quintiliano RPS, Scárdua Silva L, Yasuda CL, Cendes F, Farias AS, Damasceno A. Case report: Granzyme-B expression by T- and B- cells during severe AQP4-positive Neuromyelitis Optica spectrum disorder with fatal venous thromboembolism outcome. Front Neurol 2023; 14:1208977. [PMID: 37662034 PMCID: PMC10470460 DOI: 10.3389/fneur.2023.1208977] [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: 04/20/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Background The expression of serine protease granzyme-B (GzmB) by circulating CD8+ T lymphocytes has been recently suggested as a biomarker for poor immunotherapy response and severe disability in patients with Neuromyelitis Optica spectrum disorders (NMOSD). In parallel, venous thromboembolism (VTE) has been reported mainly in NMOSD patients exhibiting transverse myelitis. Case presentation Here, we describe an Aquaporin-4 positive (AQP4-positive) NMOSD patient who showed short myelitis (SM) and experienced a fatal pulmonary thromboembolism/lower extremity deep vein thrombosis during anti-CD20 treatment. Flow cytometry analyses from the peripheral blood revealed an enhanced cytotoxic behavior through circulating CD8+GzmB+ T, CD4+GzmB+ T lymphocytes, and residual CD19+GzmB+ B cells. Conclusions Fatal VTE may be a rare outcome, particularly in patients exhibiting SM, and may share poorly understood immunological mechanisms with AQP4-positive NMOSD severity.
Collapse
Affiliation(s)
- Vinícius Oliveira Boldrini
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| | - Mariana Rabelo Brito
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| | - Raphael Patrício Silva Quintiliano
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Lucas Scárdua Silva
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| | - Clarissa Lin Yasuda
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| | - Fernando Cendes
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| | - Alessandro Santos Farias
- Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
- Autoimmune Research Laboratory, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Alfredo Damasceno
- Neuroimaging Laboratory, Department of Neurology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
16
|
Sagan SA, Moinfar Z, Moseley CE, Dandekar R, Spencer CM, Verkman AS, Ottersen OP, Sobel RA, Sidney J, Sette A, Anderson MS, Steinman L, Wilson MR, Sabatino JJ, Zamvil SS. T cell deletional tolerance restricts AQP4 but not MOG CNS autoimmunity. Proc Natl Acad Sci U S A 2023; 120:e2306572120. [PMID: 37463205 PMCID: PMC10372680 DOI: 10.1073/pnas.2306572120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Aquaporin-4 (AQP4)-specific Th17 cells are thought to have a central role in neuromyelitis optica (NMO) pathogenesis. When modeling NMO, only AQP4-reactive Th17 cells from AQP4-deficient (AQP4-/-), but not wild-type (WT) mice, caused CNS autoimmunity in recipient WT mice, indicating that a tightly regulated mechanism normally ensures tolerance to AQP4. Here, we found that pathogenic AQP4 T cell epitopes bind MHC II with exceptionally high affinity. Examination of T cell receptor (TCR) α/β usage revealed that AQP4-specific T cells from AQP4-/- mice employed a distinct TCR repertoire and exhibited clonal expansion. Selective thymic AQP4 deficiency did not fully restore AQP4-reactive T cells, demonstrating that thymic negative selection alone did not account for AQP4-specific tolerance in WT mice. Indeed, AQP4-specific Th17 cells caused paralysis in recipient WT or B cell-deficient mice, which was followed by complete recovery that was associated with apoptosis of donor T cells. However, donor AQP4-reactive T cells survived and caused persistent paralysis in recipient mice deficient in both T and B cells or mice lacking T cells only. Thus, AQP4 CNS autoimmunity was limited by T cell-dependent deletion of AQP4-reactive T cells. In contrast, myelin oligodendrocyte glycoprotein (MOG)-specific T cells survived and caused sustained disease in WT mice. These findings underscore the importance of peripheral T cell deletional tolerance to AQP4, which may be relevant to understanding the balance of AQP4-reactive T cells in health and in NMO. T cell tolerance to AQP4, expressed in multiple tissues, is distinct from tolerance to MOG, an autoantigen restricted in its expression.
Collapse
Affiliation(s)
- Sharon A Sagan
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Zahra Moinfar
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Carson E Moseley
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Ravi Dandekar
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Collin M Spencer
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Alan S Verkman
- Department of Medicine, University of California, San Francisco, CA 94143
- Department of Physiology, University of California, San Francisco, CA 94143
| | - Ole Petter Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0316, Norway
| | - Raymond A Sobel
- Department of Pathology, Stanford University School of Medicine, Palo Alto VA Health Care System, Palo Alto, CA 94305
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Mark S Anderson
- Program in Immunology, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Michael R Wilson
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Joseph J Sabatino
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| |
Collapse
|
17
|
Kinoshita M, Okuno T. Autoimmune-mediated astrocytopathy. Inflamm Regen 2023; 43:39. [PMID: 37461118 DOI: 10.1186/s41232-023-00291-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Recently accumulating evidence identified the disease entity where astrocytes residing within the central nervous system (CNS) are the target of autoantibody-mediated autoimmunity. Aquaporin4 (AQP4) is the most common antigen to serve as astrocyte-targeted autoimmune responses. Here, in this review, the clinical and pathological aspects of AQP4-mediated astrocyte disease are discussed together with the pathogenic role of anti-AQP4 antibody. More recently, the mechanism of immune dysregulation resulting in the production of astrocyte-targeted autoantibody is also revealed, and the postulated hypothesis is discussed.
Collapse
Affiliation(s)
- Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
18
|
Shen X. Research progress on pathogenesis and clinical treatment of neuromyelitis optica spectrum disorders (NMOSDs). Clin Neurol Neurosurg 2023; 231:107850. [PMID: 37390569 DOI: 10.1016/j.clineuro.2023.107850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 04/11/2023] [Accepted: 06/23/2023] [Indexed: 07/02/2023]
Abstract
Neuromyelitis optica spectrum disorders (NMOSDs) are characteristically referred to as various central nervous system (CNS)-based inflammatory and astrocytopathic disorders, often manifested by the axonal damage and immune-mediated demyelination targeting optic nerves and the spinal cord. This review article presents a detailed view of the etiology, pathogenesis, and prescribed treatment options for NMOSD therapy. Initially, we present the epidemiology of NMOSDs, highlighting the geographical and ethnical differences in the incidence and prevalence rates of NMOSDs. Further, the etiology and pathogenesis of NMOSDs are emphasized, providing discussions relevant to various genetic, environmental, and immune-related factors. Finally, the applied treatment strategies for curing NMOSD are discussed, exploring the perspectives for developing emergent innovative treatment strategies.
Collapse
Affiliation(s)
- Xinyu Shen
- Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200000, PR China.
| |
Collapse
|
19
|
Manin A, Justo ME, Leoni J, Paz ML, Villa AM. C5a complement levels in clinical remission AQP4-IgG-positive NMO patients. Acta Neurol Belg 2023:10.1007/s13760-023-02261-7. [PMID: 37024715 DOI: 10.1007/s13760-023-02261-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Neuromyelitis Optica Spectrum Disorders (NMOSD) is an antibody-mediated disorder of the Central Nervous System where a leading role of the complement system has been demonstrated. OBJECTIVE To measure the levels of complement factors C3, C4 and C5a in serum and plasma of clinical remission patients with AQP4-IgG + NMOSD. METHODS Twelve patients with NMOSD AQP4 + according to 2015 criteria from a General Hospital in Buenos Aires, Argentina, were included in the study, and 19 age- and sex-matched healthy volunteers as a control group (HC). AQP4 antibodies were measured in serum by CBA analysis. Fresh blood samples were centrifuged to obtain serum and plasma. C3, C4, and AQP4 antibodies were measured in the serum, whereas C5a was measured in the plasma, which was obtained using Futhan (BD FUT-175®, BD Biosciences, San Jose, CA, USA). RESULTS The complement factors, C3, C4, and C5a were measured in all samples. The mean concentration of C3 was 130.7 mg/dl (SD 16.1 mg/dl), and the mean concentration of C4 was 21.6 mg/dl (SD 4.8 mg/dl); both values were within the normal reference range (C3: 84-193 mg/dl; C4: 20-40 mg/dl) and were not significantly different (p > 0.05) from the mean levels in healthy controls (C3: 116.9 mg/dl; C4: 21.9 mg/dl). When analyzing the mean plasma level of C5a, we found a statistically significant difference (p = 0.0444) between the mean concentration of C5a in NMOSD patients (43.1 ng/ml; SD 48.7 ng/ml) and the HC group (17.7 ng/ml; SD 16.7 ng/ ml). CONCLUSIONS In conclusion, the present study demonstrates that plasma C5a may be interesting to investigate as a potential biomarker of disease activity in NMOSD, in a larger and prospective cohort.
Collapse
Affiliation(s)
- Analisa Manin
- División Neurología, Hospital Gral. de Agudos Dr. José María Ramos Mejía, Bs. As, Argentina, Centro Argentino de Neuroinmunología (CADENI), Facultad de Medicina, Universidad de Buenos Aires, CABA, Cuidad Autónoma de Buenos Aires, Argentina.
| | - Mariano E Justo
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, CABA, Universidad de Buenos Aires, Cuidad Autónoma de Buenos Aires, Argentina
- Facultad de Farmacia y Bioquímica, CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), CABA, Cuidad Autónoma de Buenos Aires, Argentina
| | - Juliana Leoni
- Facultad de Farmacia y Bioquímica, CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), CABA, Cuidad Autónoma de Buenos Aires, Argentina
| | - Mariela L Paz
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, CABA, Universidad de Buenos Aires, Cuidad Autónoma de Buenos Aires, Argentina
- Facultad de Farmacia y Bioquímica, CONICET-Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), CABA, Cuidad Autónoma de Buenos Aires, Argentina
| | - Andrés M Villa
- División Neurología, Hospital Gral. de Agudos Dr. José María Ramos Mejía, Bs. As, Argentina, Centro Argentino de Neuroinmunología (CADENI), Facultad de Medicina, Universidad de Buenos Aires, CABA, Cuidad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
20
|
Zakani M, Nigritinou M, Ponleitner M, Takai Y, Hofmann D, Hillebrand S, Höftberger R, Bauer J, Lasztoczi B, Misu T, Kasprian G, Rommer P, Bradl M. Paths to hippocampal damage in neuromyelitis optica spectrum disorders. Neuropathol Appl Neurobiol 2023; 49:e12893. [PMID: 36811295 PMCID: PMC10947283 DOI: 10.1111/nan.12893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Abstract
AIMS Many patients with neuromyelitis optica spectrum disorders (NMOSD) suffer from cognitive impairment affecting memory, processing speed and attention and suffer from depressive symptoms. Because some of these manifestations could trace back to the hippocampus, several magnetic resonance imaging (MRI) studies have been performed in the past, with a number of groups describing volume loss of the hippocampus in NMOSD patients, whereas others did not observe such changes. Here, we addressed these discrepancies. METHODS We performed pathological and MRI studies on the hippocampi of NMOSD patients, combined with detailed immunohistochemical analysis of hippocampi from experimental models of NMOSD. RESULTS We identified different pathological scenarios for hippocampal damage in NMOSD and its experimental models. In the first case, the hippocampus was compromised by the initiation of astrocyte injury in this brain region and subsequent local effects of microglial activation and neuronal damage. In the second case, loss of hippocampal volume was seen by MRI in patients with large tissue-destructive lesions in the optic nerves or the spinal cord, and the pathological work-up of tissue derived from a patient with such lesions revealed subsequent retrograde neuronal degeneration affecting different axonal tracts and neuronal networks. It remains to be seen whether remote lesions and associated retrograde neuronal degeneration on their own are sufficient to cause extensive volume loss of the hippocampus, or whether they act in concert with small astrocyte-destructive, microglia-activating lesions in the hippocampus that escape detection by MRI, either due to their small size or due to the chosen time window for examination. CONCLUSIONS Different pathological scenarios can culminate in hippocampal volume loss in NMOSD patients.
Collapse
Affiliation(s)
- Mona Zakani
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Magdalini Nigritinou
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | | | - Yoshiki Takai
- Department of NeurologyTohoku University Graduate School of MedicineSendaiJapan
| | - Daniel Hofmann
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Sophie Hillebrand
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Romana Höftberger
- Department of Neurology, Division of Neuropathology and NeurochemistryMedical University of ViennaViennaAustria
| | - Jan Bauer
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Balint Lasztoczi
- Division of Cognitive Neurobiology, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Tatsuro Misu
- Department of NeurologyTohoku University Graduate School of MedicineSendaiJapan
| | - Gregor Kasprian
- Division of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Paulus Rommer
- Department of NeurologyMedical University of ViennaViennaAustria
| | - Monika Bradl
- Division of Neuroimmunology, Center for Brain ResearchMedical University of ViennaViennaAustria
| |
Collapse
|
21
|
Mader S, Ho S, Wong HK, Baier S, Winklmeier S, Riemer C, Rübsamen H, Fernandez IM, Gerhards R, Du C, Chuquisana O, Lünemann JD, Lux A, Nimmerjahn F, Bradl M, Kawakami N, Meinl E. Dissection of complement and Fc-receptor-mediated pathomechanisms of autoantibodies to myelin oligodendrocyte glycoprotein. Proc Natl Acad Sci U S A 2023; 120:e2300648120. [PMID: 36943883 PMCID: PMC10068779 DOI: 10.1073/pnas.2300648120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/08/2023] [Indexed: 03/23/2023] Open
Abstract
Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) have recently been established to define a new disease entity, MOG-antibody-associated disease (MOGAD), which is clinically overlapping with multiple sclerosis. MOG-specific antibodies (Abs) from patients are pathogenic, but the precise effector mechanisms are currently still unknown and no therapy is approved for MOGAD. Here, we determined the contributions of complement and Fc-receptor (FcR)-mediated effects in the pathogenicity of MOG-Abs. Starting from a recombinant anti-MOG (mAb) with human IgG1 Fc, we established MOG-specific mutant mAbs with differential FcR and C1q binding. We then applied selected mutants of this MOG-mAb in two animal models of experimental autoimmune encephalomyelitis. First, we found MOG-mAb-induced demyelination was mediated by both complement and FcRs about equally. Second, we found that MOG-Abs enhanced activation of cognate MOG-specific T cells in the central nervous system (CNS), which was dependent on FcR-, but not C1q-binding. The identification of complement-dependent and -independent pathomechanisms of MOG-Abs has implications for therapeutic strategies in MOGAD.
Collapse
Affiliation(s)
- Simone Mader
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Samantha Ho
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
- Graduate School of Systemic Neuroscience, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Hoi Kiu Wong
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Selia Baier
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Carolina Riemer
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
| | - Heike Rübsamen
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Iris Marti Fernandez
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Ramona Gerhards
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Cuilian Du
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Omar Chuquisana
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149Münster, Germany
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149Münster, Germany
| | - Anja Lux
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nürnberg, 91058Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen91058, Germany
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, 1090Vienna, Austria
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg-Martinsried, Germany
| |
Collapse
|
22
|
Yick LW, Ma OKF, Chan EYY, Yau KX, Kwan JSC, Chan KH. T follicular helper cells contribute to pathophysiology in a model of neuromyelitis optica spectrum disorders. JCI Insight 2023; 8:161003. [PMID: 36649074 PMCID: PMC9977492 DOI: 10.1172/jci.insight.161003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are inflammatory autoimmune disorders of the CNS. IgG autoantibodies targeting the aquaporin-4 water channel (AQP4-IgGs) are the pathogenic effector of NMOSD. Dysregulated T follicular helper (Tfh) cells have been implicated in loss of B cell tolerance in autoimmune diseases. The contribution of Tfh cells to disease activity and therapeutic potential of targeting these cells in NMOSD remain unclear. Here, we established an autoimmune model of NMOSD by immunizing mice against AQP4 via in vivo electroporation. After AQP4 immunization, mice displayed AQP4 autoantibodies in blood circulation, blood-brain barrier disruption, and IgG infiltration in spinal cord parenchyma. Moreover, AQP4 immunization induced motor impairments and NMOSD-like pathologies, including astrocytopathy, demyelination, axonal loss, and microglia activation. These were associated with increased splenic Tfh, Th1, and Th17 cells; memory B cells; and plasma cells. Aqp4-deficient mice did not display motor impairments and NMOSD-like pathologies after AQP4 immunization. Importantly, abrogating ICOS/ICOS-L signaling using anti-ICOS-L antibody depleted Tfh cells and suppressed the response of Th1 and Th17 cells, memory B cells, and plasma cells in AQP4-immunized mice. These findings were associated with ameliorated motor impairments and spinal cord pathologies. This study suggests a role of Tfh cells in the pathophysiology of NMOSD in a mouse model with AQP4 autoimmunity and provides an animal model for investigating the immunological mechanisms underlying AQP4 autoimmunity and developing therapeutic interventions targeting autoimmune reactions in NMOSD.
Collapse
|
23
|
Kanbayashi T, Ogawa G, Ito T, Hokkoku K, Oishi C, Hatanaka Y, Sonoo M. Utility of the tibial nerve somatosensory evoked potentials in differentiating between neuromyelitis optica spectrum disorders and multiple sclerosis. Mult Scler Relat Disord 2023; 70:104503. [PMID: 36610361 DOI: 10.1016/j.msard.2023.104503] [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: 09/08/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
BACKGROUND Somatosensory evoked potentials (SEPs) are widely used for the diagnosis and evaluation of neuromyelitis optica spectrum disorder (NMOSD) and multiple sclerosis (MS). However, whether the parameters of tibial nerve SEPs can help to distinguish NMOSD from MS remains unclear. Thus, the aim of this study was to investigate the utility of tibial nerve SEP parameters in differentiating patients with NMOSD and MS. METHODS The clinical data of patients with NMOSD or MS treated in our institution between 2005 and 2021 were retrospectively extracted from our electronic database. Additional inclusion criteria were presentation with sensory symptoms in the lower extremities with corresponding lesions in the magnetic resonance images as well as available data on anti-aquaporin-4 antibodies and tibial nerve SEPs. The Z-scores of the N21-P38 interval (central sensory conduction time), P38 latency, and P38 amplitude were compared between the patients with NMOSD and MS. The relationship of disease severity with the parameters of the tibial nerve SEPs was also evaluated. RESULTS Twenty patients with NMOSD and 13 patients with MS were enrolled. The Z-scores of the N21-P38 interval and P38 latency were significantly higher in the MS group than in the NMOSD group (p < 0.05 and p < 0.01, respectively), whereas there was no difference in the Z-scores of the P38 amplitude between the two groups. In the MS group, only the N21-P38 interval and P38 latency were significantly correlated with disease severity (p < 0.05 and p < 0.01, respectively). In contrast, none of the tibial nerve SEP parameters were significantly correlated with disease severity in the NMOSD group. CONCLUSION Evaluation of the N21-P38 interval and P38 latency in tibial nerve SEPs potentially helps in differentiating between NMOSD and MS.
Collapse
Affiliation(s)
- Takamichi Kanbayashi
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| | - Go Ogawa
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| | - Tatsuya Ito
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| | - Keiichi Hokkoku
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| | - Chizuko Oishi
- Department of Neurology, Faculty of Medicine, Kyorin University, Tokyo, Japan.
| | - Yuki Hatanaka
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| | - Masahiro Sonoo
- Department of Neurology, Teikyo University School of Medicine, Kaga 2-11-1, Tokyo 1738605, Japan.
| |
Collapse
|
24
|
Azam T, Bukhari SH, Liaqat U, Miran W. Emerging Methods in Biosensing of Immunoglobin G-A Review. SENSORS (BASEL, SWITZERLAND) 2023; 23:676. [PMID: 36679468 PMCID: PMC9862834 DOI: 10.3390/s23020676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
Human antibodies are produced due to the activation of immune system components upon exposure to an external agent or antigen. Human antibody G, or immunoglobin G (IgG), accounts for 75% of total serum antibody content. IgG controls several infections by eradicating disease-causing pathogens from the body through complementary interactions with toxins. Additionally, IgG is an important diagnostic tool for certain pathological conditions, such as autoimmune hepatitis, hepatitis B virus (HBV), chickenpox and MMR (measles, mumps, and rubella), and coronavirus-induced disease 19 (COVID-19). As an important biomarker, IgG has sparked interest in conducting research to produce robust, sensitive, selective, and economical biosensors for its detection. To date, researchers have used different strategies and explored various materials from macro- to nanoscale to be used in IgG biosensing. In this review, emerging biosensors for IgG detection have been reviewed along with their detection limits, especially electrochemical biosensors that, when coupled with nanomaterials, can help to achieve the characteristics of a reliable IgG biosensor. Furthermore, this review can assist scientists in developing strategies for future research not only for IgG biosensors but also for the development of other biosensing systems for diverse targets.
Collapse
Affiliation(s)
- Tehmina Azam
- School of Chemical and Materials Engineering (SCME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Syed Hassan Bukhari
- College of Computational Sciences and Natural Sciences, Minerva University, San Francisco, CA 94103, USA
| | - Usman Liaqat
- School of Chemical and Materials Engineering (SCME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| | - Waheed Miran
- School of Chemical and Materials Engineering (SCME), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan
| |
Collapse
|
25
|
Lerch M, Schanda K, Lafon E, Würzner R, Mariotto S, Dinoto A, Wendel EM, Lechner C, Hegen H, Rostásy K, Berger T, Wilflingseder D, Höftberger R, Reindl M. More Efficient Complement Activation by Anti–Aquaporin-4 Compared With Anti–Myelin Oligodendrocyte Glycoprotein Antibodies. NEUROLOGY - NEUROIMMUNOLOGY NEUROINFLAMMATION 2023; 10:10/1/e200059. [DOI: 10.1212/nxi.0000000000200059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/19/2022] [Indexed: 11/23/2022]
Abstract
Background and ObjectivesThe objective was to study complement-mediated cytotoxicity induced by immunoglobulin G (IgG) anti–aquaporin-4 antibodies (AQP4-IgG) and anti–myelin oligodendrocyte glycoprotein antibodies (MOG-IgG) in human serum samples from patients suffering from the rare demyelinating diseases of the CNS neuromyelitis optica spectrum disorder (NMOSD) and MOG-IgG–associated disease (MOGAD).MethodsA cell-based assay with HEK293A cells expressing different MOG isoforms (MOGα1-3β1-3) or AQP4-M23 was used. Cells were incubated with human MOG-IgG or AQP4-IgG–positive serum samples together with active or heat-inactivated human complement, and complement-dependent cytotoxicity (CDC) was measured with a lactate dehydrogenase assay. To further quantify antibody-mediated cell damage, formation of the terminal complement complex (TCC) was analyzed by flow cytometry. In addition, immunocytochemistry of the TCC and complement component 3 (C3) was performed.ResultsAQP4-IgG–positive serum samples induced higher CDC and TCC levels than MOG-IgG–positive sera. Notably, both showed a correlation between antibody titers and CDC and also between titers and TCC levels. In addition, all 6 MOG isoforms tested (MOGα1-3β1-3) could induce at least some CDC; however, the strongest MOG-IgG–induced CDC levels were found on MOGα1, MOGα3, and MOGβ1. Different MOG-IgG binding patterns regarding recognition of different MOG isoforms were investigated, and it was found that MOG-IgG recognizing all 6 isoforms again induced highest CDC levels on MOGα1and MOGβ1. Furthermore, surface staining of TCC and C3 revealed positive staining on all 6 MOG isoforms tested, as well as on AQP4-M23.DiscussionBoth MOG-IgG and AQP4-IgG are able to induce CDC in a titer-dependent manner. However, AQP4-IgG showed markedly higher levels of CDC compared with MOG in vitro on target cells. This further highlights the role of complement in AQP4-IgG–mediated disease and diminishes the importance of complement activation in MOG-IgG–mediated autoimmune disease.
Collapse
|
26
|
Yamamoto M, Okuno T, Piao JL, Shimizu M, Miyamoto K, Nukui T, Kinoshita M, Koda T, Dini Haryuni R, Mochizuki H, Sugimoto T, Nakatsuji Y. Identification of double-stranded DNA in the cerebrospinal fluid of patients with acute neuromyelitis optica spectrum disorder. J Clin Neurosci 2023; 107:129-132. [PMID: 36543044 DOI: 10.1016/j.jocn.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/28/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory disease of the central nervous system (CNS) characterized by severe myelitis and optic neuritis. Double-stranded DNA (dsDNA) is involved in the pathogenesis of various autoimmune diseases, such as systemic lupus erythematosus. However, its role in NMOSD remains unclear. In this study, the concentration of dsDNA in the cerebrospinal fluid (CSF) was quantified in 23 patients with NMOSD and 16 patients with other neurological diseases (ONDs). CSF dsDNA levels in patients with NMOSD (median: 0.03 ng/µL) were significantly higher than those in patients with ONDs (median: 0.01 ng/μl). CSF dsDNA levels showed no significant difference before and after treatment. Elevation of CSF dsDNA levels may suggest its essential role in the augmentation of CNS inflammation in patients with NMOSD.
Collapse
Affiliation(s)
- Mamoru Yamamoto
- Department of Neurology, Faculty of Medicine, University of Toyama, Japan
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, Japan.
| | - Jin-Lan Piao
- Department of Neurology, Faculty of Medicine, University of Toyama, Japan
| | - Mikito Shimizu
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | | | - Takamasa Nukui
- Department of Neurology, Faculty of Medicine, University of Toyama, Japan
| | - Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Toru Koda
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | - Ratna Dini Haryuni
- Department of Neurology, Faculty of Medicine, University of Toyama, Japan; Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency, Indonesia
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Japan
| | | | - Yuji Nakatsuji
- Department of Neurology, Faculty of Medicine, University of Toyama, Japan.
| |
Collapse
|
27
|
Schindler P, Aktas O, Ringelstein M, Wildemann B, Jarius S, Paul F, Ruprecht K. Glial fibrillary acidic protein as a biomarker in neuromyelitis optica spectrum disorder: a current review. Expert Rev Clin Immunol 2023; 19:71-91. [PMID: 36378751 DOI: 10.1080/1744666x.2023.2148657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing, often debilitating neuroinflammatory disease, whose predominant clinical manifestations are longitudinally extensive transverse myelitis and optic neuritis. About 80% of the patients with an NMOSD phenotype have pathogenic autoantibodies against the astrocyte water channel aquaporin-4 (AQP4-IgG). While therapeutic options for NMOSD have greatly expanded in recent years, well-established biomarkers for prognosis or treatment response are still lacking. Glial fibrillary acidic protein (GFAP) is mainly expressed in astrocytes and can be detected in cerebrospinal fluid (CSF) and blood of patients with NMOSD. AREAS COVERED Here, we comprehensively review the current knowledge on GFAP as a biomarker in NMOSD. EXPERT OPINION In patients with AQP4-IgG+ NMOSD, GFAP levels are elevated in CSF and serum during acute attacks and correlate with disability, consistent with the pathophysiology of this antibody-mediated astrocytopathy. Serum GFAP levels tend to be higher in AQP4-IgG+ NMOSD than in its differential diagnoses, multiple sclerosis, and myelin oligodendrocyte antibody-associated disease. Importantly, serum GFAP levels in AQP4-IgG+ NMOSD during remission may be predictive of future disease activity. Serial serum GFAP measurements are emerging as a biomarker to monitor disease activity in AQP4-IgG+ NMOSD and could have the potential for application in clinical practice.
Collapse
Affiliation(s)
- Patrick Schindler
- Experimental and Clinical Research Center, A Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Marius Ringelstein
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Neurology, Center for Neurology and Neuropsychiatry, LVR-Klinikum, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, A Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
28
|
Höftberger R, Lassmann H, Berger T, Reindl M. Pathogenic autoantibodies in multiple sclerosis - from a simple idea to a complex concept. Nat Rev Neurol 2022; 18:681-688. [PMID: 35970870 DOI: 10.1038/s41582-022-00700-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
The role of autoantibodies in multiple sclerosis (MS) has been enigmatic since the first description, many decades ago, of intrathecal immunoglobulin production in people with this condition. Some studies have indicated that MS pathology is heterogeneous, with an antibody-associated subtype - characterized by B cells (in varying quantities), antibodies and complement - existing alongside other subtypes with different pathologies. However, subsequent evidence suggested that some cases originally diagnosed as MS with autoantibody-mediated demyelination were more likely to be neuromyelitis optica spectrum disorder or myelin oligodendrocyte glycoprotein antibody-associated disease. These findings raise the important question of whether an autoantibody-mediated MS subtype exists and whether pathogenic MS-associated autoantibodies remain to be identified. Potential roles of autoantibodies in MS could range from specific antibodies defining the disease to a non-disease-specific amplification of cellular immune responses and other pathophysiological processes. In this Perspective, we review studies that have attempted to identify MS-associated autoantibodies and provide our opinions on their possible roles in the pathophysiology of MS.
Collapse
Affiliation(s)
- Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
29
|
Kalluri SR, Srivastava R, Kenet S, Tanti GK, Dornmair K, Bennett JL, Misgeld T, Hemmer B, Wyss MT, Herwerth M. P2R Inhibitors Prevent Antibody-Mediated Complement Activation in an Animal Model of Neuromyelitis Optica : P2R Inhibitors Prevent Autoantibody Injury. Neurotherapeutics 2022; 19:1603-1616. [PMID: 35821382 PMCID: PMC9606199 DOI: 10.1007/s13311-022-01269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 11/28/2022] Open
Abstract
Purinergic 2 receptors (P2Rs) contribute to disease-related immune cell signaling and are upregulated in various pathological settings, including neuroinflammation. P2R inhibitors have been used to treat inflammatory diseases and can protect against complement-mediated cell injury. However, the mechanisms behind these anti-inflammatory properties of P2R inhibitors are not well understood, and their potential in CNS autoimmunity is underexplored. Here, we tested the effects of P2R inhibitors on glial toxicity in a mouse model of neuromyelitis optica spectrum disorder (NMOSD). NMOSD is a destructive CNS autoimmune disorder, in which autoantibodies against astrocytic surface antigen Aquaporin 4 (AQP4) mediate complement-dependent loss of astrocytes. Using two-photon microscopy in vivo, we found that various classes of P2R inhibitors prevented AQP4-IgG/complement-dependent astrocyte death. In vitro, these drugs inhibited the binding of AQP4-IgG or MOG-IgG to their antigen in a dose-dependent manner. Size-exclusion chromatography and circular dichroism spectroscopy revealed a partial unfolding of antibodies in the presence of various P2R inhibitors, suggesting a shared interference with IgG antibodies leading to their conformational change. Our study demonstrates that P2R inhibitors can disrupt complement activation by direct interaction with IgG. This mechanism is likely to influence the role of P2R inhibitors in autoimmune disease models and their therapeutic impact in human disease.
Collapse
Affiliation(s)
- Sudhakar Reddy Kalluri
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Rajneesh Srivastava
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Selin Kenet
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Goutam K Tanti
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, LMU Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Colorado, USA
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Matthias T Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University Zurich and ETH Zurich, Zurich, Switzerland
| | - Marina Herwerth
- Department of Neurology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany.
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Stathopoulos P, Dalakas MC. The role of complement and complement therapeutics in neuromyelitis optica spectrum disorders. Expert Rev Clin Immunol 2022; 18:933-945. [PMID: 35899480 DOI: 10.1080/1744666x.2022.2105205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorders (NMOSD) are characterized in the majority of cases by the presence of IgG1 autoantibodies against aquaporin 4 (AQP4) and myelin-oligodendrocyte glycoprotein (MOG), both capable of activating complement. AREAS COVERED We review evidence of complement involvement in NMOSD pathophysiology from pathological, in vitro, in vivo, human studies, and clinical trials. EXPERT OPINION In AQP4 NMOSD, complement deposition is a prominent pathological feature, while in vitro and in vivo studies have demonstrated complement-dependent pathogenicity of AQP4 antibodies. Consistent with these studies, the anti-C5 monoclonal antibody eculizumab was remarkably effective and safe in a phase 2/3 trial of AQP4-NMOSD patents leading to FDA-approved indication. Several other anti-complement agents, either approved or in trials for other neuro-autoimmunities, like myasthenia, CIDP, and GBS, are also relevant to NMOSD generating an exciting group of evolving immunotherapies. Limited but compelling in vivo and in vitro data suggest that anti-complement therapeutics may be also applicable to a subset of MOG NMOSD patients with severe disease. Overall, anticomplement agents, along with the already approved anti-IL6 and anti-CD19 monoclonal antibodies sartralizumab and inebilizumab, are rapidly changing the therapeutic algorithm in NMOSD, a previously difficult-to-treat autoimmune neurological disorder.
Collapse
Affiliation(s)
- Panos Stathopoulos
- Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
31
|
Neuromyelitis Optica Spectrum Disorder: From Basic Research to Clinical Perspectives. Int J Mol Sci 2022; 23:ijms23147908. [PMID: 35887254 PMCID: PMC9323454 DOI: 10.3390/ijms23147908] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 02/05/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory disease of the central nervous system characterized by relapses and autoimmunity caused by antibodies against the astrocyte water channel protein aquaporin-4. Over the past decade, there have been significant advances in the biologic knowledge of NMOSD, which resulted in the IDENTIFICATION of variable disease phenotypes, biomarkers, and complex inflammatory cascades involved in disease pathogenesis. Ongoing clinical trials are looking at new treatments targeting NMOSD relapses. This review aims to provide an update on recent studies regarding issues related to NMOSD, including the pathophysiology of the disease, the potential use of serum and cerebrospinal fluid cytokines as disease biomarkers, the clinical utilization of ocular coherence tomography, and the comparison of different animal models of NMOSD.
Collapse
|
32
|
Lin L, Wu Y, Hang H, Lu J, Ding Y. Plasma Complement 3 and Complement 4 Are Promising Biomarkers for Distinguishing NMOSD From MOGAD and Are Associated With the Blood-Brain-Barrier Disruption in NMOSD. Front Immunol 2022; 13:853891. [PMID: 35898513 PMCID: PMC9309329 DOI: 10.3389/fimmu.2022.853891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background and ObjectiveNeuromyelitis optica spectrum disorders (NMOSD) and myelin oligodendrocyte glycoprotein antibody (MOG-IgG) associated disease (MOGAD) are autoimmune inflammatory demyelinating diseases of the central nervous system (CNS). As the clinical features of NMOSD are similar to MOGAD, diagnostic confusion exists between the two diseases. To better discriminate NMOSD from MOGAD, we investigated whether the plasma levels of complement 3 (C3) and complement 4 (C4) are different in NMOSD and MOGAD during the acute attacks of the diseases. We sought to determine whether C3 or C4 has an influence on the features of NMOSD.MethodsIn this observational study, data from 73 aquaporin-4 antibodies (AQP4-IgG) positive NMOSD patients and 22 MOG-IgG positive MOGAD patients were collected retrospectively. Demographics, clinical characteristics, plasma parameters, and cerebrospinal fluid (CSF) findings will be analyzed for comparability between the two groups. Immunoglobulin-G (IgG) and albumin were measured in both plasma and CSF. Plasma levels of C3 and C4 were measured and compared between the NMOSD, MOGAD, and 42 healthy controls (HC). The correlations between plasma C3, C4, and NMOSD clinical parameters were analyzed.ResultsThe ages of onset were later in the AQP4-IgG positive NMOSD group and females predominated, which differed from the MOGAD group, whose ages were younger and with a slight male preponderance. The AQP4-IgG positive NMOSD patients presented with the clinical symptoms of optic neuritis (ON) and transverse myelitis (TM), whereas encephalitis symptoms were more prevalent in MOGAD patients. CSF analysis shows that slight but not significantly higher white cell count (WCC) and protein were observed in the MOGAD group than in the AQP4-IgG positive NMOSD group. The plasma levels of IgG in MOGAD patients are significantly lower (p = 0.027) than in NMOSD patients. On the contrary, the plasma levels of albumin in MOGAD were higher than in NMOSD, which reached statistical significance (p = 0.039). Both the plasma C3 and C4 levels in the NMOSD group were significantly lower than in MOGAD and HC. The receiver operating characteristic (ROC) curve of the prediction model comprises C3 and C4 to distinguish NMOSD from MOGAD [area under the curve (AUC): 0.731, 0.645], which are considered to have discriminatory values. The results of Spearman’s analysis revealed that there was a significant positive correlation between the plasma C3 and the CSF WCC (r = 0.383, p = 0.040). There was an inverse correlation between plasma C4 and plasma IgG (r = -0.244, p = 0.038). Plasma C3 or C4 was significantly positively correlated with CSF albumin and Q-Alb, which is considered a measure of blood-brain barrier (BBB) disruption.ConclusionDuring the acute phase of NMOSD and MOGAD, plasma C3 and C4 may become potential biomarkers for distinguishing the two diseases and reflecting the NMOSD BBB damage.
Collapse
|
33
|
Zveik O, Rechtman A, Haham N, Adini I, Canello T, Lavon I, Brill L, Vaknin-Dembinsky A. Sera of Neuromyelitis Optica Patients Increase BID-Mediated Apoptosis in Astrocytes. Int J Mol Sci 2022; 23:ijms23137117. [PMID: 35806122 PMCID: PMC9266359 DOI: 10.3390/ijms23137117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Neuromyelitis optica (NMO) is a rare disease usually presenting with bilateral or unilateral optic neuritis with simultaneous or sequential transverse myelitis. Autoantibodies directed against aquaporin-4 (AQP4-IgG) are found in most patients. They are believed to cross the blood−brain barrier, target astrocytes, activate complement, and eventually lead to astrocyte destruction, demyelination, and axonal damage. However, it is still not clear what the primary pathological event is. We hypothesize that the interaction of AQP4-IgG and astrocytes leads to DNA damage and apoptosis. We studied the effect of sera from seropositive NMO patients and healthy controls (HCs) on astrocytes’ immune gene expression and viability. We found that sera from seropositive NMO patients led to higher expression of apoptosis-related genes, including BH3-interacting domain death agonist (BID), which is the most significant differentiating gene (p < 0.0001), and triggered more apoptosis in astrocytes compared to sera from HCs. Furthermore, NMO sera increased DNA damage and led to a higher expression of immunological genes that interact with BID (TLR4 and NOD-1). Our findings suggest that sera of seropositive NMO patients might cause astrocytic DNA damage and apoptosis. It may be one of the mechanisms implicated in the primary pathological event in NMO and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Omri Zveik
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ariel Rechtman
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Nitzan Haham
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Irit Adini
- Department of Surgery, Harvard Medical School, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA;
| | - Tamar Canello
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Iris Lavon
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Livnat Brill
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Correspondence: ; Tel.: +972-2-677-7741
| |
Collapse
|
34
|
Wang Y, Zhang J, Chang H, Wang H, Xu W, Cong H, Zhang X, Liu J, Yin L. NMO-IgG induce interleukin-6 release via activation of the NF-κB signaling pathway in astrocytes. Neuroscience 2022; 496:96-104. [PMID: 35659638 DOI: 10.1016/j.neuroscience.2022.05.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelinating disorder of the central nervous system (CNS) that frequently affects the optic nerve and spinal cord. Interleukin-6 (IL-6) is considered a key cytokine in the pathogenesis of NMOSD, and the level of IL-6 is significantly increased in the sera and cerebrospinal fluid (CSF) of patients with NMOSD. We have reported that the production of IL-6 depends on the JAK/STAT3 signaling pathway. However, it is not clear whether the NF-κB-dependent inflammatory response stimulated by neuromyelitis optica IgG (NMO-IgG) could also drive the production of IL-6 in astrocytes. In this study, we used an in vitro model of primary rat astrocytes stimulated by NMO-IgG to study the role of the NF-κB signaling pathway in mediating the release of IL-6. First, we confirmed that the level of IL-6 was significantly higher in the sera of NMOSD patients than that of healthy people by humoral fluid analysis and that NMO-IgG can significantly induce the release of IL-6 from astrocytes by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. Then, Western blotting and immunocytochemistry showed that NMO-IgG can activate the intracellular NF-κB signaling pathway. Finally, it was found that S3633, an inhibitor of the NF-κB signaling pathway, can effectively inhibit the increase in IL-6 levels. These results prove that the production of IL-6 is partly mediated by the NF-κB signaling pathway, providing a potential effective strategy for targeted treatment of NMOSD.
Collapse
Affiliation(s)
- Yupeng Wang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Jingwen Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Haoxiao Chang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Huabing Wang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Wangshu Xu
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Hengri Cong
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Xinghu Zhang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China
| | - Jianghong Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing 100053, China; Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| | - Linlin Yin
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Capital Medical University, No.119 South 4thRing West Road, Fengtai District, Beijing 100160, China; Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
35
|
Olude MA, Mouihate A, Mustapha OA, Farina C, Quintana FJ, Olopade JO. Astrocytes and Microglia in Stress-Induced Neuroinflammation: The African Perspective. Front Immunol 2022; 13:795089. [PMID: 35707531 PMCID: PMC9190229 DOI: 10.3389/fimmu.2022.795089] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Africa is laden with a youthful population, vast mineral resources and rich fauna. However, decades of unfortunate historical, sociocultural and leadership challenges make the continent a hotspot for poverty, indoor and outdoor pollutants with attendant stress factors such as violence, malnutrition, infectious outbreaks and psychological perturbations. The burden of these stressors initiate neuroinflammatory responses but the pattern and mechanisms of glial activation in these scenarios are yet to be properly elucidated. Africa is therefore most vulnerable to neurological stressors when placed against a backdrop of demographics that favor explosive childbearing, a vast population of unemployed youths making up a projected 42% of global youth population by 2030, repressive sociocultural policies towards women, poor access to healthcare, malnutrition, rapid urbanization, climate change and pollution. Early life stress, whether physical or psychological, induces neuroinflammatory response in developing nervous system and consequently leads to the emergence of mental health problems during adulthood. Brain inflammatory response is driven largely by inflammatory mediators released by glial cells; namely astrocytes and microglia. These inflammatory mediators alter the developmental trajectory of fetal and neonatal brain and results in long-lasting maladaptive behaviors and cognitive deficits. This review seeks to highlight the patterns and mechanisms of stressors such as poverty, developmental stress, environmental pollutions as well as malnutrition stress on astrocytes and microglia in neuroinflammation within the African context.
Collapse
Affiliation(s)
- Matthew Ayokunle Olude
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
- *Correspondence: Matthew Ayokunle Olude,
| | - Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Health Sciences Centre, Kuwait University, Kuwait City, Kuwait
| | - Oluwaseun Ahmed Mustapha
- Vertebrate Morphology, Environmental Toxicology and Neuroscience Unit, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - Cinthia Farina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) San Raffaele Scientific Institute, Institute of Experimental Neurology (INSPE) and Division of Neuroscience, Milan, Italy
| | - Francisco Javier Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - James Olukayode Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
36
|
Immuno-pathogenesis of neuromyelitis optica and emerging therapies. Semin Immunopathol 2022; 44:599-610. [DOI: 10.1007/s00281-022-00941-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/20/2022] [Indexed: 01/01/2023]
|
37
|
Nagahata K, Suzuki S, Yokochi R, Nei Y, Hagino N. Recurrent Optic Perineuritis With Myelin Oligodendrocyte Glycoprotein Antibody-Associated Disease Complicated With Granulomatous Polyangiitis. Cureus 2022; 14:e25239. [PMID: 35747030 PMCID: PMC9215109 DOI: 10.7759/cureus.25239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 12/01/2022] Open
Abstract
Optic perineuritis (OPN) is an intraorbital inflammatory disease that targets the optic nerve sheath, which can cause severe vision loss. OPN has been recently reported to be sometimes caused by myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease (MOGAD). MOGAD is rarely reported to be complicated with other autoimmune diseases. We report the first rare case of MOG-associated OPN complicated with granulomatous with polyangiitis (GPA). The vision loss, in this case, was initially considered to be caused by cavernous sinusitis in GPA. However, she was diagnosed with MOGAD with serial MRI findings and positive MOG antibody and had been successfully treated with glucocorticoid and tocilizumab for one and half years. This case emphasized the importance of evaluating the MOG antibody in a patient with recurrent OPN, complicated with vasculitis.
Collapse
|
38
|
Mader S, Brimberg L, Vo A, Strohl JJ, Crawford JM, Bonnin A, Carrión J, Campbell D, Huerta TS, La Bella A, Berlin R, Dewey SL, Hellman M, Eidelberg D, Dujmovic I, Drulovic J, Bennett JL, Volpe BT, Huerta PT, Diamond B. In utero exposure to maternal anti-aquaporin-4 antibodies alters brain vasculature and neural dynamics in male mouse offspring. Sci Transl Med 2022; 14:eabe9726. [PMID: 35442708 PMCID: PMC9973562 DOI: 10.1126/scitranslmed.abe9726] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The fetal brain is constantly exposed to maternal IgG before the formation of an effective blood-brain barrier (BBB). Here, we studied the consequences of fetal brain exposure to an antibody to the astrocytic protein aquaporin-4 (AQP4-IgG) in mice. AQP4-IgG was cloned from a patient with neuromyelitis optica spectrum disorder (NMOSD), an autoimmune disease that can affect women of childbearing age. We found that embryonic radial glia cells in neocortex express AQP4. These cells are critical for blood vessel and BBB formation through modulation of the WNT signaling pathway. Male fetuses exposed to AQP4-IgG had abnormal cortical vasculature and lower expression of WNT signaling molecules Wnt5a and Wnt7a. Positron emission tomography of adult male mice exposed in utero to AQP4-IgG revealed increased blood flow and BBB leakiness in the entorhinal cortex. Adult male mice exposed in utero to AQP4-IgG had abnormal cortical vessels, fewer dendritic spines in pyramidal and stellate neurons, and more S100β+ astrocytes in the entorhinal cortex. Behaviorally, they showed impairments in the object-place memory task. Neural recordings indicated that their grid cell system, within the medial entorhinal cortex, did not map the local environment appropriately. Collectively, these data implicate in utero binding of AQP4-IgG to radial glia cells as a mechanism for alterations of the developing male brain and adds NMOSD to the conditions in which maternal IgG may cause persistent brain dysfunction in offspring.
Collapse
Affiliation(s)
- Simone Mader
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
- Institute of Clinical Neuroimmunology, Biomedical Center of the Ludwig Maximilian University of Munich, Munich 82152, Germany
| | - Lior Brimberg
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - An Vo
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Joshua J. Strohl
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - James M. Crawford
- Department of Pathology and Laboratory Medicine, Northwell Health, Manhasset, NY 11030, USA
| | - Alexandre Bonnin
- Department of Physiology and Neurosciences, Zilkha Neurogenetic Institute, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Joseph Carrión
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Delcora Campbell
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Tomás S. Huerta
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Andrea La Bella
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Roseann Berlin
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Stephen L. Dewey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Matthew Hellman
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - David Eidelberg
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Irena Dujmovic
- Clinical Center of Serbia University School of Medicine, Belgrade, 11000, Serbia
- Department of Neurology, University of North Carolina, School of Medicine, Chapel Hill, NC 27517, USA
| | - Jelena Drulovic
- Clinical Center of Serbia University School of Medicine, Belgrade, 11000, Serbia
| | - Jeffrey L. Bennett
- Department of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Denver, School of Medicine, Denver, CO 80045, USA
| | - Bruce T. Volpe
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| | - Patricio T. Huerta
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Betty Diamond
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset NY 11030, USA
| |
Collapse
|
39
|
Abe Y, Yasui M. Aquaporin-4 in Neuromyelitis Optica Spectrum Disorders: A Target of Autoimmunity in the Central Nervous System. Biomolecules 2022; 12:biom12040591. [PMID: 35454180 PMCID: PMC9030581 DOI: 10.3390/biom12040591] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/20/2022] Open
Abstract
Since the discovery of a specific autoantibody in patients with neuromyelitis optica spectrum disorder (NMOSD) in 2004, the water channel aquaporin-4 (AQP4) has attracted attention as a target of autoimmune diseases of the central nervous system. In NMOSD, the autoantibody (NMO-IgG) binds to the extracellular loops of AQP4 as expressed in perivascular astrocytic end-feet and disrupts astrocytes in a complement-dependent manner. NMO-IgG is an excellent marker for distinguishing the disease from other inflammatory demyelinating diseases, such as multiple sclerosis. The unique higher-order structure of AQP4—called orthogonal arrays of particles (OAPs)—as well as its subcellular localization may play a crucial role in the pathogenesis of the disease. Recent studies have also demonstrated complement-independent cytotoxic effects of NMO-IgG. Antibody-induced endocytosis of AQP4 has been suggested to be involved in this mechanism. This review focuses on the binding properties of antibodies that recognize the extracellular region of AQP4 and the characteristics of AQP4 that are implicated in the pathogenesis of NMOSD.
Collapse
Affiliation(s)
- Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Keio University Global Research Institute, Tokyo 108-8345, Japan
- Correspondence: (Y.A.); (M.Y.); Tel.: +81-3-5363-3751 (M.Y.)
| | - Masato Yasui
- Department of Pharmacology, Keio University School of Medicine, Tokyo 160-8582, Japan
- Keio University Global Research Institute, Tokyo 108-8345, Japan
- Correspondence: (Y.A.); (M.Y.); Tel.: +81-3-5363-3751 (M.Y.)
| |
Collapse
|
40
|
Lassmann H. The Contribution of Neuropathology to Multiple Sclerosis Research. Eur J Neurol 2022; 29:2869-2877. [PMID: 35427431 PMCID: PMC9544263 DOI: 10.1111/ene.15360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Hans Lassmann
- Center for Brain Research Medical University of Vienna Austria
| |
Collapse
|
41
|
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: 3.7] [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
|
42
|
Matsumoto Y, Ohyama A, Kubota T, Ikeda K, Kaneko K, Takai Y, Warita H, Takahashi T, Misu T, Aoki M. MOG Antibody-Associated Disorders Following SARS-CoV-2 Vaccination: A Case Report and Literature Review. Front Neurol 2022; 13:845755. [PMID: 35299613 PMCID: PMC8922017 DOI: 10.3389/fneur.2022.845755] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/24/2022] [Indexed: 01/28/2023] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG) antibody-associated disorder (MOGAD) is a newly identified autoimmune demyelinating disorder that is often associated with acute disseminated encephalomyelitis and usually occurs postinfection or postvaccination. Here we report a case of MOGAD after mRNA severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination. A previously healthy 68-year-old woman presented to our department with gradually worsening numbness on the right side of her face, which began 14 days after her second dose of an mRNA-1273 vaccination. The patient's brain MRI revealed a right cerebellar peduncle lesion with gadolinium enhancement, a typical finding of MOGAD. A neurological examination revealed paresthesia on her right V2 and V3 areas. Other neurological examinations were unremarkable. Laboratory workups were positive for serum MOG-IgG as assessed by live cell-based assays and the presence of oligoclonal bands in the cerebrospinal fluid (CSF). The patient's serum test results for cytoplasmic-antineutrophil cytoplasmic antibodies, perinuclear-cytoplasmic-antineutrophil cytoplasmic antibodies, GQ1b-antibodies, and aquaporin-4 antibodies (AQP4-IgG) were all negative. Tests for soluble interleukin (IL)-2 receptors in the serum, IL-6 in the CSF and skin pricks, and angiotensin converting enzyme tests were all unremarkable. The patient was diagnosed with MOGAD after receiving an mRNA SARS-CoV-2 vaccination. After two courses of intravenous methylprednisolone treatment, the patient's symptoms improved and her cerebellar peduncle lesion shrunk slightly without gadolinium enhancement. To date, there have only been two cases of monophasic MOGAD following SARS-CoV-2 vaccination, including both the ChAdOx1 nCOV-19 and mRNA-1273 vaccines, and the prognosis is generally similar to other typical MOGAD cases. Although the appearance of MOG antibodies is relatively rare in post-COVID-19-vaccine demyelinating diseases, MOGAD should be considered in patients with central nervous system (CNS) demyelinating diseases after receiving a SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Yuki Matsumoto
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ayane Ohyama
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Takafumi Kubota
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Kensuke Ikeda
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Kimihiko Kaneko
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Hitoshi Warita
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Toshiyuki Takahashi
- Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Hospital, Sendai, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Neurology, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
43
|
Clarke L, Bukhari W, O'Gorman CM, Khalilidehkordi E, Arnett S, Woodhall M, Prain KM, Parratt JDE, Barnett MH, Marriott MP, McCombe PA, Sutton I, Boggild M, Brownlee W, Carroll WM, Hodgkinson S, Macdonell RAL, Mason DF, Pereira J, Slee M, Das C, Henderson APD, Kermode AG, Lechner-Scott J, Waters P, Sun J, Broadley SA. Response to treatment in NMOSD: the Australasian experience. Mult Scler Relat Disord 2022; 58:103408. [PMID: 35216788 DOI: 10.1016/j.msard.2021.103408] [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: 09/30/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is associated with significant morbidity and mortality. Several therapies have been recommended for NMOSD and more recently clinical trials have demonstrated efficacy for three monoclonal antibody therapies. We present a retrospective observational study of treatment response in NMOSD. METHODS This was a retrospective, unblinded, observational study of treatment efficacy for rituximab and traditional immunosuppressive therapy in patients with AQP4 antibody positive NMOSD. Treatment efficacy was assessed using annualised relapse rates (ARR), time to first relapse and expanded disability status scale (EDSS) scores. RESULTS Complete relapse and treatment data were available for 43/68 (63%) of AQP4 antibody positive NMOSD cases covering 74 episodes of treatment. In a time to first relapse analysis rituximab showed a risk ratio of 0.23 (95% CI 0.08 - 0.65) when compared with no treatment and there was a non-significant reduction in ARR of 35% compared to pre-treatment. β-interferon (p = 0.0002) and cyclophosphamide (p = 0.0034) were associated with an increased ARR compared to pre-treatment. Rituximab (median 4.0 [range 0.0 - 7.0]; p = 0.042) and traditional immunosuppressive therapy (median 4.0 [range 0.0 - 8.0]; p = 0.016) were associated with a lower final EDSS compared to β-interferon (median 6.0 [range 4.0 - 7.5]). CONCLUSIONS These data provide additional support for the use of rituximab in preference to traditional immunosuppressive agents and MS disease modifying therapies as first line treatment of NMOSD.
Collapse
Affiliation(s)
- Laura Clarke
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; Department of Neurology Princess Alexandra Hospital, Woolloongabba QLD 4102, Australia
| | - Wajih Bukhari
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; St Vincent's Hospital Melbourne, Fitzroy VIC 3065, AustraliA
| | - Cullen M O'Gorman
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; Department of Neurology Princess Alexandra Hospital, Woolloongabba QLD 4102, Australia; Department of Neurology, Mater Hospital Brisbane, South Brisbane QLD, 4101, Australia
| | - Elham Khalilidehkordi
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Herston QLD 4029, Australia
| | - Simon Arnett
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; Department of Neurology, Gold Coast University Hospital, Southport QLD 4215, Australia
| | - Mark Woodhall
- Nuffield Department of Clinical Neurosciences, John Radcliffe Infirmary, University of Oxford, Oxford OX3 9DU, UK
| | - Kerri M Prain
- Department of Immunology, Pathology Queensland, Royal Brisbane and Women's Hospital, Herston QLD 4006, Australia
| | - John D E Parratt
- Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown NSW 2006, Australia
| | - Michael H Barnett
- Brain and Mind Research Institute, University of Sydney, Camperdown NSW 2006, Australia
| | - Mark P Marriott
- Melbourne Brain Centre, Royal Melbourne Hospital, University of Melbourne, Parkville VIC 3052, Australia
| | - Pamela A McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, Herston QLD 4029, Australia; Centre for Clinical Research, Royal Brisbane and Women's Hospital, University of Queensland, Herston QLD 4029, AustraliA
| | - Ian Sutton
- Department of Neurology, St Vincent's Hospital, Darlinghurst NSW 2010, Australia
| | - Mike Boggild
- Department of Neurology, Townsville Hospital, Douglas QLD 4814, Australia
| | - Wallace Brownlee
- Department of Neurology, Auckland City Hospital, Grafton 1023, New Zealand; Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK
| | - William M Carroll
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, Queen Elizabeth II Medical Centre, University of Western Australia, Nedlands WA 6009, AustraliA
| | - Suzanne Hodgkinson
- South Western Sydney Medical School, Liverpool Hospital, University of New South Wales, Liverpool NSW 2170, Australia
| | | | - Deborah F Mason
- Department of Neurology, Christchurch Hospital, Christchurch 8140, New Zealand
| | - Jennifer Pereira
- Department of Neurology, Auckland City Hospital, Grafton 1023, New Zealand
| | - Mark Slee
- Flinders Medical Centre, Flinders University, Bedford Park SA 5042, Australia
| | - Chandi Das
- Department of Neurology, Canberra Hospital, Garran ACT 2605, Australia
| | | | - Allan G Kermode
- Centre for Neuromuscular and Neurological Disorders, Perron Institute for Neurological and Translational Science, Queen Elizabeth II Medical Centre, University of Western Australia, Nedlands WA 6009, AustraliA; Institute for Immunology and Infectious Disease, Murdoch University, Murdoch WA 6150, AustraliA
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, University of Newcastle, New Lambton Heights NSW 2305, AustralIA
| | | | - Patrick Waters
- Department of Neurology, Gold Coast University Hospital, Southport QLD 4215, Australia
| | - Jing Sun
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia
| | - Simon A Broadley
- Menzies Health Institute Queensland, Gold Coast Campus, Griffith University QLD 4222, Australia; Department of Neurology, Gold Coast University Hospital, Southport QLD 4215, Australia.
| |
Collapse
|
44
|
Hoshino Y, Noto D, Sano S, Tomizawa Y, Yokoyama K, Hattori N, Miyake S. Dysregulated B cell differentiation towards antibody-secreting cells in neuromyelitis optica spectrum disorder. J Neuroinflammation 2022; 19:6. [PMID: 34991631 PMCID: PMC8740356 DOI: 10.1186/s12974-021-02375-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/28/2021] [Indexed: 11/24/2022] Open
Abstract
Background Anti-aquaporin 4 (AQP4) antibody (AQP4-Ab) is involved in the pathogenesis of neuromyelitis optica spectrum disorder (NMOSD). However, the mechanism involved in AQP4-Ab production remains unclear. Methods We analyzed the immunophenotypes of patients with NMOSD and other neuroinflammatory diseases as well as healthy controls (HC) using flow cytometry. Transcriptome analysis of B cell subsets obtained from NMOSD patients and HCs was performed. The differentiation capacity of B cell subsets into antibody-secreting cells was analyzed. Results The frequencies of switched memory B (SMB) cells and plasmablasts were increased and that of naïve B cells was decreased in NMOSD patients compared with relapsing–remitting multiple sclerosis patients and HC. SMB cells from NMOSD patients had an enhanced potential to differentiate into antibody-secreting cells when cocultured with T peripheral helper cells. Transcriptome analysis revealed that the profiles of B cell lineage transcription factors in NMOSD were skewed towards antibody-secreting cells and that IL-2 signaling was upregulated, particularly in naïve B cells. Naïve B cells expressing CD25, a receptor of IL-2, were increased in NMOSD patients and had a higher potential to differentiate into antibody-secreting cells, suggesting CD25+ naïve B cells are committed to differentiate into antibody-secreting cells. Conclusions To the best of our knowledge, this is the first study to demonstrate that B cells in NMOSD patients are abnormally skewed towards antibody-secreting cells at the transcriptome level during the early differentiation phase, and that IL-2 might participate in this pathogenic process. Our study indicates that CD25+ naïve B cells are a novel candidate precursor of antibody-secreting cells in autoimmune diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02375-w.
Collapse
Affiliation(s)
- Yasunobu Hoshino
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.,Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Shuhei Sano
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yuji Tomizawa
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazumasa Yokoyama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
45
|
Tong Y, Liu J, Yang T, Wang J, Zhao T, Kang Y, Fan Y. Association of Pain with Plasma C5a in Patients with Neuromyelitis Optica Spectrum Disorders During Remission. Neuropsychiatr Dis Treat 2022; 18:1039-1046. [PMID: 35615424 PMCID: PMC9124695 DOI: 10.2147/ndt.s359620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To investigate the association of pain with plasma C5a levels and other related inflammatory cytokines in neuromyelitis optica spectrum disorders (NMOSD) patients during remission. PARTICIPANTS AND METHODS NMOSD patients (n = 87) and healthy controls (HC; n = 44) were consecutively recruited between January 2017 and April 2018. Plasma complement 5 (C5), C5a, interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-1β levels were detected. Visual Analogue Scale (VAS), ID pain scale, 24-item Hamilton Depression Scale (HAMD), Multiple Sclerosis Impact Scale (MSIS-29), and Kurtzke Expanded Disability Status Scale (EDSS) were used to evaluate the degree and types of pain, the existence of depression and anxiety, and the life quality and disability status of patients. Binary logistic regression equation was used to assess the association of pain with plasma C5a levels. RESULTS Among the 87 NMOSD patients, 40 complained of pain that in 67.5% (27/40) of cases had a neuropathic component (ID pain ≥2). Plasma C5a, IL-6, TNF-α, and IL-1β levels were significantly elevated in NMOSD patients than in HC. Plasma C5 levels were negatively correlated with the time from sampling to the last relapse or disease onset. NMOSD patients with pain had higher plasma C5a levels, and they suffered from a higher disability, more anxiety, and worse life quality compared to those patients without pain. In NMOSD patients with pain, there were not significant differences between plasma levels of C5, C5a, IL-6, TNF-α, or IL-1β, regardless of neuropathic pain or not. Binary logistic regression showed that the OR of plasma C5a level was 1.002, with gender and EDSS score were identified as independent factors associated with pain in NMOSD. CONCLUSION NMOSD patients during remission had elevated C5a and related inflammatory cytokines levels in peripheral blood. Elevated C5a may have a unique role in the pathogenesis of pain in NMOSD patients.
Collapse
Affiliation(s)
- Yanping Tong
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,Beijing Integrative Medicine on Encephalopathy Research Institution, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Jie Liu
- Department of Respiratory Medicine, Beijing First Hospital of Integrated Chinese and Western Medicine, Beijing, 100026, People's Republic of China
| | - Tao Yang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,Beijing Integrative Medicine on Encephalopathy Research Institution, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Jingwen Wang
- Department of Neurology, Beijing Miyun District Hospital of Traditional Chinese Medicine, Beijing, 101599, People's Republic of China
| | - Tianyou Zhao
- Department of Neurology, Beijing Miyun District Hospital of Traditional Chinese Medicine, Beijing, 101599, People's Republic of China
| | - Yuezhi Kang
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,Beijing Integrative Medicine on Encephalopathy Research Institution, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| | - Yongping Fan
- Department of Traditional Chinese Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China.,Beijing Integrative Medicine on Encephalopathy Research Institution, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, People's Republic of China
| |
Collapse
|
46
|
Fu CC, Gao C, Zhang HH, Mao YQ, Lu JQ, Petritis B, Huang AS, Yang XG, Long YM, Huang RP. Serum molecular biomarkers in neuromyelitis optica and multiple sclerosis. Mult Scler Relat Disord 2022; 59:103527. [DOI: 10.1016/j.msard.2022.103527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 12/18/2022]
|
47
|
Yang H, Liu W, Wu YF, Zhu DS, Shen XF, Guan YT. Lymphocyte Subsets Are Associated with Disease Status in Neuromyelitis Optica Spectrum Disorders. Neuroimmunomodulation 2022; 29:296-305. [PMID: 34903694 DOI: 10.1159/000520745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/28/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE At present, studies on lymphocytes are mostly conducted on CD19+ B cells and CD27+ B cells in neuromyelitis optica spectrum disorders (NMOSDs), but the exact changes in lymphocyte subsets (CD19+ B cells, CD3+ T cells, CD4+ Th cells, CD8+ Ts cells, the CD4+/CD8+ ratio, and NK [CD56+ CD16] cells) have rarely been studied. This study aimed to assess lymphocyte subset changes in patients with NMOSD. METHODS We performed a cross-sectional study of consecutive patients with acute NMOSD (n = 41), chronic NMOSD (n = 21), and healthy individuals (n = 44). Peripheral blood samples were obtained upon admission, and lymphocyte subsets were analyzed by flow cytometry. Levels of lymphocyte subsets among 3 groups were compared and its correlation with the length of spinal cord lesions was analyzed. RESULTS The levels of peripheral blood CD19+ B cells were significantly higher in patients with acute and chronic NMOSD than in healthy controls (HCs) (17.91 ± 8.7%, 13.08 ± 7.562%, and 12.48 ± 3.575%, respectively; p < 0.001) and were positively correlated with the length of spinal cord lesions in acute NMOSD (r = 0.433, p < 0.05). The peripheral blood CD4+/CD8+ ratio was significantly lower in patients with acute NMOSD and chronic NMOSD than in HCs (1.497 ± 0.6387, 1.33 ± 0.5574, and 1.753 ± 0.659, respectively; p < 0.05), and the levels of peripheral blood NK (CD56+ CD16) cells were significantly lower in patients with acute and chronic NMOSD than in HCs (13.6 ± 10.13, 11.11 ± 7.057, and 14.7 [interquartile range = 9.28], respectively; p < 0.01). CONCLUSIONS The levels of certain subsets of peripheral blood lymphocytes are associated with disease status in NMOSD.
Collapse
Affiliation(s)
- Hong Yang
- Department of Neurology, The First Rehabilitation Hospital of Shanghai, School of Medicine, Tongji University, Shanghai, China,
| | - Wei Liu
- Department of Neurology, The First Rehabilitation Hospital of Shanghai, School of Medicine, Tongji University, Shanghai, China
| | - Yi-Fan Wu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - De-Sheng Zhu
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xia-Feng Shen
- Department of Neurology, The First Rehabilitation Hospital of Shanghai, School of Medicine, Tongji University, Shanghai, China
| | - Yang-Tai Guan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
48
|
Park J, Kim CH. Regulation of common neurological disorders by gut microbial metabolites. Exp Mol Med 2021; 53:1821-1833. [PMID: 34857900 PMCID: PMC8741890 DOI: 10.1038/s12276-021-00703-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/06/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The gut is connected to the CNS by immunological mediators, lymphocytes, neurotransmitters, microbes and microbial metabolites. A mounting body of evidence indicates that the microbiome exerts significant effects on immune cells and CNS cells. These effects frequently result in the suppression or exacerbation of inflammatory responses, the latter of which can lead to severe tissue damage, altered synapse formation and disrupted maintenance of the CNS. Herein, we review recent progress in research on the microbial regulation of CNS diseases with a focus on major gut microbial metabolites, such as short-chain fatty acids, tryptophan metabolites, and secondary bile acids. Pathological changes in the CNS are associated with dysbiosis and altered levels of microbial metabolites, which can further exacerbate various neurological disorders. The cellular and molecular mechanisms by which these gut microbial metabolites regulate inflammatory diseases in the CNS are discussed. We highlight the similarities and differences in the impact on four major CNS diseases, i.e., multiple sclerosis, Parkinson's disease, Alzheimer's disease, and autism spectrum disorder, to identify common cellular and molecular networks governing the regulation of cellular constituents and pathogenesis in the CNS by microbial metabolites.
Collapse
Affiliation(s)
- Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
- Mary H. Weiser Food Allergy Center, Center for Gastrointestinal Research, and Rogel Center for Cancer Research, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
49
|
Pache F, Ringelstein M, Aktas O, Kleiter I, Jarius S, Siebert N, Bellmann-Strobl J, Paul F, Ruprecht K. C3 and C4 complement levels in AQP4-IgG-positive NMOSD and in MOGAD. J Neuroimmunol 2021; 360:577699. [PMID: 34464830 DOI: 10.1016/j.jneuroim.2021.577699] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 08/08/2021] [Accepted: 08/21/2021] [Indexed: 11/30/2022]
Abstract
While complement-dependent cytotoxicity (CDC) is a known effector mechanism in aquaporin-4-immunoglobulin (Ig)G-positive (AQP4-IgG+) neuromyelitis optica spectrum disorder (NMOSD), the role of CDC in myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) is less clear. We determined complement C3 and C4 plasma concentrations in patients with clinically stable AQP4-IgG+ NMOSD (n = 16), MOGAD (n = 15), early multiple sclerosis (MS, n = 19) and in healthy controls (HC, n = 18). C4 was lower in AQP4-IgG+ NMOSD than in MOGAD, MS and HC (p < 0.05, pairwise comparisons). C3 was lower in AQP4-IgG+ NMOSD than in MS (p = 0.034). These findings suggest subtle complement consumption in clinically stable AQP4-IgG+ NMOSD, but not in MOGAD.
Collapse
Affiliation(s)
- Florence Pache
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology, Berlin, Germany
| | - Marius Ringelstein
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Neurology, Center for Neurology and Neuropsychiatry, LVR-Klinikum Düsseldorf, Düsseldorf, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Ingo Kleiter
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, Bochum, Germany; Marianne-Strauß-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke gGmbH, Berg, Germany
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Nadja Siebert
- 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, Charité - Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany; Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Judith Bellmann-Strobl
- 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, Charité - Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany; Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Friedemann Paul
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology, Berlin, Germany; 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, Charité - Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine, Berlin-Buch, Germany
| | - Klemens Ruprecht
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology, Berlin, Germany.
| |
Collapse
|
50
|
Guo Y, Lennon VA, Parisi JE, Popescu B, Vasquez C, Pittock SJ, Howe CL, Lucchinetti CF. Spectrum of sublytic astrocytopathy in neuromyelitis optica. Brain 2021; 145:1379-1390. [PMID: 34718426 PMCID: PMC9128820 DOI: 10.1093/brain/awab394] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/22/2021] [Accepted: 09/18/2021] [Indexed: 11/30/2022] Open
Abstract
Neuromyelitis optica is an autoimmune inflammatory disorder targeting aquaporin-4 water channels in CNS astrocytes. Histopathological descriptions of astrocytic lesions reported in neuromyelitis optica so far have emphasized a characteristic loss of aquaporin-4, with deposition of IgG and complement and lysis of astrocytes, but sublytic reactions have been underappreciated. We performed a multi-modality study of 23 neuromyelitis optica autopsy cases (clinically and/or pathologically confirmed; 337 tissue blocks). By evaluating astrocytic morphology, immunohistochemistry and AQP4 RNA transcripts, and their associations with demyelinating activity, we documented a spectrum of astrocytopathy in addition to complement deposition, microglial reaction, granulocyte infiltration and regenerating activity. Within advanced demyelinating lesions, and in periplaque areas, there was remarkable hypertrophic astrogliosis, more subtle than astrocytic lysis. A degenerative component was suggested by ‘dystrophic’ morphology, cytoplasmic vacuolation, Rosenthal fibres and associated stress protein markers. The abundance of AQP4 mRNA transcripts in sublytic reactive astrocytes devoid of aquaporin-4 protein supported in vivo restoration following IgG-induced aquaporin-4 endocytosis/degradation. Astrocytic alterations extending beyond demyelinating lesions speak to astrocytopathy being an early and primary event in the evolving neuromyelitis optica lesion. Focal astrocytopathy observed without aquaporin-4 loss or lytic complement component deposition verifies that astrocytic reactions in neuromyelitis optica are not solely dependent on IgG-mediated aquaporin-4 loss or lysis by complement or by IgG-dependent leucocyte mediators. We conclude that neuromyelitis optica reflects a global astrocytopathy, initiated by binding of IgG to aquaporin-4 and not simply definable by demyelination and astrocytic lysis. The spectrum of astrocytic morphological changes in neuromyelitis optica attests to the complexity of factors influencing the range of astrocytic physiological responses to a targeted attack by aquaporin-4-specific IgG. Sublytic astrocytic reactions are no doubt an important determinant of the lesion’s evolution and potential for repair. Pharmacological manipulation of the astrocytic stress response may offer new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Yong Guo
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Vanda A Lennon
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Joseph E Parisi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Bogdan Popescu
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | - Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Claudia F Lucchinetti
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|