1
|
Chen S, Zhang Y, Liu K, Xiao Z, Liu X, Xu Z, Qiu W, Xu Y, Tang C, Yang H. Inhibition of C3/C3aR Pathway Alleviates Visual and Optic Nerve Impairments in a Rat Model of Neuromyelitis Optica Spectrum Disorder. Eur J Neurosci 2025; 61:e70103. [PMID: 40261058 DOI: 10.1111/ejn.70103] [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: 12/21/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
Accumulating evidence suggests that the complement C3/C3a receptor (C3aR) pathway plays a pivotal role in the pathogenesis of neurodegenerative and autoimmune disorders, particularly neuromyelitis optica spectrum disorders (NMOSDs). Despite this, the specific pathogenic effect and mechanism of C3/C3aR pathway in NMOSD remains elusive. In this study, we demonstrated for the first time that the expression of C3aR and its upstream ligand C3 is significantly upregulated in the optic nerve of a rat model of NMOSD-related optic neuritis (NMOSD-ON). Our findings reveal that this upregulation leads to blood-brain barrier (BBB) disruption, demyelination and neuronal damage. Through the use of a novel C3aR inhibitor, JR14a, we demonstrate its effectiveness in reducing C3aR expression and mitigating pathological damage in the optic nerve. Furthermore, our transcriptome sequencing analysis of NMOSD optic nerve tissues reveals extensive enrichment of inflammation- and immune response-related pathways, with particular emphasis on the complement and coagulation cascades pathway. This study not only elucidated the crucial role of the C3-C3aR pathway in NMOSD-ON pathogenesis but also provided a new promising therapeutic target for NMOSD through C3aR pathway inhibition.
Collapse
Affiliation(s)
- Siqi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Yurong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Kaiqun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Zhiqiang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Xiaoning Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Ziyan Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ying Xu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hui Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual Science, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
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
|
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
|
Bigotte M, Groh AMR, Marignier R, Stratton JA. Pathogenic role of autoantibodies at the ependyma in autoimmune disorders of the central nervous system. Front Cell Neurosci 2023; 17:1257000. [PMID: 37771929 PMCID: PMC10525373 DOI: 10.3389/fncel.2023.1257000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
Ependymal cells make up the epithelial monolayer that lines the brain ventricles and the spinal cord central canal that are filled with cerebrospinal fluid. The ependyma has several functions, including regulating solute exchange between the cerebrospinal fluid and parenchyma, controlling microcirculation of cerebrospinal fluid via coordinated ciliary beating, and acting as a partial barrier. Dysregulation of these functions can lead to waste clearance impairment, cerebrospinal fluid accumulation, hydrocephalus, and more. A role for ependymal cells in a variety of neurological disorders has been proposed, including in neuromyelitis optica and multiple sclerosis, two autoimmune demyelinating diseases of the central nervous system, where periventricular damage is common. What is not known is the mechanisms behind how ependymal cells become dysregulated in these diseases. In neuromyelitis optica, it is well established that autoantibodies directed against Aquaporin-4 are drivers of disease, and it has been shown recently that these autoantibodies can drive ependymal cell dysregulation. We propose a similar mechanism is at play in multiple sclerosis, where autoantibodies targeting a glial cell protein called GlialCAM on ependymal cells are contributing to disease. GlialCAM shares high molecular similarities with the Epstein-Barr virus (EBV) protein EBNA1. EBV has recently been shown to be necessary for multiple sclerosis initiation, yet how EBV mediates pathogenesis, especially in the periventricular area, remains elusive. In this perspective article, we discuss how ependymal cells could be targeted by antibody-related autoimmune mechanisms in autoimmune demyelinating diseases and how this is implicated in ventricular/periventricular pathology.
Collapse
Affiliation(s)
- Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Adam M. R. Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Romain Marignier
- Forgetting Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, Bron, France
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuroinflammation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Bron, France
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
Thangaleela S, Sivamaruthi BS, Radha A, Kesika P, Chaiyasut C. Neuromyelitis Optica Spectrum Disorders: Clinical Perspectives, Molecular Mechanisms, and Treatments. APPLIED SCIENCES 2023; 13:5029. [DOI: 10.3390/app13085029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Neuromyelitis optica (NMO) is a rare autoimmune inflammatory disorder affecting the central nervous system (CNS), specifically the optic nerve and the spinal cord, with severe clinical manifestations, including optic neuritis (ON) and transverse myelitis. Initially, NMO was wrongly understood as a condition related to multiple sclerosis (MS), due to a few similar clinical and radiological features, until the discovery of the AQP4 antibody (NMO-IgG/AQP4-ab). Various etiological factors, such as genetic-environmental factors, medication, low levels of vitamins, and others, contribute to the initiation of NMO pathogenesis. The autoantibodies against AQP4 target the AQP4 channel at the blood–brain barrier (BBB) of the astrocyte end feet, which leads to high permeability or leakage of the BBB that causes more influx of AQP4-antibodies into the cerebrospinal fluid (CSF) of NMO patients. The binding of AQP4-IgG onto the AQP4 extracellular epitopes initiates astrocyte damage through complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC). Thus, a membrane attack complex is formed due to complement cascade activation; the membrane attack complex targets the AQP4 channels in the astrocytes, leading to astrocyte cell damage, demyelination of neurons and oligodendrocytes, and neuroinflammation. The treatment of NMOSD could improve relapse symptoms, restore neurological functions, and alleviate immunosuppression. Corticosteroids, apheresis therapies, immunosuppressive drugs, and B cell inactivating and complement cascade blocking agents have been used to treat NMOSD. This review intends to provide all possible recent studies related to molecular mechanisms, clinical perspectives, and treatment methodologies of the disease, particularly focusing on recent developments in clinical criteria and therapeutic formulations.
Collapse
Affiliation(s)
- Subramanian Thangaleela
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Arumugam Radha
- Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, India
| | - Periyanaina Kesika
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
6
|
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
|
7
|
Bigotte M, Gimenez M, Gavoille A, Deligiannopoulou A, El Hajj A, Croze S, Goumaidi A, Malleret G, Salin P, Giraudon P, Ruiz A, Marignier R. Ependyma: a new target for autoantibodies in neuromyelitis optica? Brain Commun 2022; 4:fcac307. [PMID: 36751497 PMCID: PMC9897195 DOI: 10.1093/braincomms/fcac307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/26/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system characterized by the presence of autoantibodies (called NMO-IgG) targeting aquaporin-4. Aquaporin-4 is expressed at the perivascular foot processes of astrocytes, in the glia limitans, but also at the ependyma. Most studies have focused on studying the pathogenicity of NMO-IgG on astrocytes, and NMO is now considered an astrocytopathy. However, periependymal lesions are observed in NMO suggesting that ependymal cells could also be targeted by NMO-IgG. Ependymal cells regulate CSF-parenchyma molecular exchanges and CSF flow, and are a niche for sub-ventricular neural stem cells. Our aim was to examine the effect of antibodies from NMO patients on ependymal cells. We exposed two models, i.e. primary cultures of rat ependymal cells and explant cultures of rat lateral ventricular wall whole mounts, to purified IgG of NMO patients (NMO-IgG) for 24 hours. We then evaluated the treatment effect using immunolabelling, functional assays, ependymal flow analysis and bulk RNA sequencing. For each experiment, the effects were compared with those of purified IgG from a healthy donors and non-treated cells. We found that: (i) NMO-IgG induced aquaporin-4 agglomeration at the surface of ependymal cells and induced cell enlargement in comparison to controls. In parallel, it induced an increase in gap junction connexin-43 plaque size; (ii) NMO-IgG altered the orientation of ciliary basal bodies and functionally impaired cilia motility; (iii) NMO-IgG activated the proliferation of sub-ventricular neural stem cells; (iv) treatment with NMO-IgG up-regulated the expression of pro-inflammatory cytokines and chemokines in the transcriptomic analysis. Our study showed that NMO-IgG can trigger an early and specific reactive phenotype in ependymal cells, with functional alterations of intercellular communication and cilia, activation of sub-ventricular stem cell proliferation and the secretion of pro-inflammatory cytokines. These findings suggest a key role for ependymal cells in the early phase of NMO lesion formation.
Collapse
Affiliation(s)
- Maxime Bigotte
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Marie Gimenez
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Antoine Gavoille
- Service de neurologie, sclérose en plaques, pathologies de la myéline et neuroinflammation, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 69500 Bron, France,Service de Biostatistique-Bioinformatique, Hospices Civils de Lyon, 69495 Pierre-Bénitem, France
| | - Adamantia Deligiannopoulou
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Aseel El Hajj
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Severine Croze
- Profilexpert, Genomic and Microgenomic Service, Claude Bernard Lyon 1 University, SFR santé LYON-EST, UCBL-INSERM US 7-CNRS UMS 3453, 69008 Lyon, France
| | | | - Gael Malleret
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Paul Salin
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Pascale Giraudon
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Anne Ruiz
- FORGETTING Team—Lyon Neuroscience Research Center, INSERM U1028, CNRS UMR 5292, Claude Bernard Lyon 1 University, 69675 Bron, France
| | - Romain Marignier
- Correspondence to: Romain Marignier Centre de référence des maladies inflammatoires rares du cerveau et de la moelle Service de neurologie, sclérose en plaques pathologies de la myéline et neuro-inflammation Hôpital Neurologique Pierre Wertheimer 59 boulevard Pinel, 69677 Bron cedex, France E-mail:
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 2.3] [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
|
9
|
Nelke C, Spatola M, Schroeter CB, Wiendl H, Lünemann JD. Neonatal Fc Receptor-Targeted Therapies in Neurology. Neurotherapeutics 2022; 19:729-740. [PMID: 34997443 PMCID: PMC9294083 DOI: 10.1007/s13311-021-01175-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
Autoantibodies are increasingly recognized for their pathogenic potential in a growing number of neurological diseases. While myasthenia gravis represents the prototypic antibody (Ab)-mediated neurological disease, many more disorders characterized by Abs targeting neuronal or glial antigens have been identified over the past two decades. Depletion of humoral immune components including immunoglobulin G (IgG) through plasma exchange or immunoadsorption is a successful therapeutic strategy in most of these disease conditions. The neonatal Fc receptor (FcRn), primarily expressed by endothelial and myeloid cells, facilitates IgG recycling and extends the half-life of IgG molecules. FcRn blockade prevents binding of endogenous IgG to FcRn, which forces these antibodies into lysosomal degradation, leading to IgG depletion. Enhancing the degradation of endogenous IgG by FcRn-targeted therapies proved to be a powerful therapeutic approach in patients with generalized MG and is currently being tested in clinical trials for several other neurological diseases including autoimmune encephalopathies, neuromyelitis optica spectrum disorders, and inflammatory neuropathies. This review illustrates mechanisms of FcRn-targeted therapies and appraises their potential to treat neurological diseases.
Collapse
Affiliation(s)
- Christopher Nelke
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | - Marianna Spatola
- MIT and Harvard Medical School, Ragon Institute of MGH, Cambridge, MA, USA
| | - Christina B Schroeter
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | - Heinz Wiendl
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Munster, Germany
| | - Jan D Lünemann
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Munster, Germany.
| |
Collapse
|
10
|
Dutta A, Das M. Deciphering the Role of Aquaporins in Metabolic Diseases: A Mini Review. Am J Med Sci 2022; 364:148-162. [DOI: 10.1016/j.amjms.2021.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 06/16/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022]
|
11
|
Parrot S, Corscadden A, Lallemant L, Benyamine H, Comte JC, Huguet-Lachon A, Gourdon G, Gomes-Pereira M. Defects in Mouse Cortical Glutamate Uptake Can Be Unveiled In Vivo by a Two-in-One Quantitative Microdialysis. ACS Chem Neurosci 2022; 13:134-142. [PMID: 34923816 DOI: 10.1021/acschemneuro.1c00634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular glutamate levels are maintained low by efficient transporters, whose dysfunction can cause neuronal hyperexcitability, excitotoxicity, and neurological disease. While many methods estimate glutamate uptake in vitro/ex vivo, a limited number of techniques address glutamate transport in vivo. Here, we used in vivo microdialysis in a two-in-one approach combining reverse dialysis of isotopic glutamate to measure uptake ability and zero-flow (ZF) methods to quantify extracellular glutamate levels. The complementarity of both techniques is discussed on methodological and anatomical basis. We used a transgenic mouse model of human disease, expressing low levels of the EAAT-2/GLT1 glutamate transporter, to validate our approach in a relevant animal model. As expected, isotopic analysis revealed an overall decrease in glutamate uptake, while the ZF method unveiled higher extracellular glutamate levels in these mice. We propose a sensitive and expedite two-in-one microdialysis approach that is sufficiently robust to reveal significant differences in neurotransmitter uptake and extracellular levels through the analysis of a relatively low number of animals.
Collapse
Affiliation(s)
- Sandrine Parrot
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Alex Corscadden
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Louison Lallemant
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Hélène Benyamine
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Jean-Christophe Comte
- Inserm U1028, CNRS UMR5292, Université de Lyon, Lyon Neuroscience Research Center, Bron F-69500, France
| | - Aline Huguet-Lachon
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Geneviève Gourdon
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Mário Gomes-Pereira
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| |
Collapse
|
12
|
Cousins O, Hodges A, Schubert J, Veronese M, Turkheimer F, Miyan J, Engelhardt B, Roncaroli F. The Blood‐CSF‐Brain Route of Neurological Disease: The Indirect Pathway into the Brain. Neuropathol Appl Neurobiol 2021; 48:e12789. [DOI: 10.1111/nan.12789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Oliver Cousins
- Department of Neuroimaging, IoPPN, King’s College London London United Kingdom
| | - Angela Hodges
- Department of Old Age Psychiatry, IoPPN, King’s College London London United Kingdom
| | - Julia Schubert
- Department of Neuroimaging, IoPPN, King’s College London London United Kingdom
| | - Mattia Veronese
- Department of Neuroimaging, IoPPN, King’s College London London United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, IoPPN, King’s College London London United Kingdom
| | - Jaleel Miyan
- Division of Neuroscience and Experimental Psychology School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, M13 9PL
| | | | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, M13 9PL
- Geoffrey Jefferson Brain Research Centre; Manchester Academic Health Science Centre Manchester UK
| |
Collapse
|
13
|
Wright SK, Wassmer E, Vincent A. Pathogenic antibodies to AQP4: Neuromyelitis optica spectrum disorder (NMOSD). BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2021; 1863:183772. [PMID: 34509490 DOI: 10.1016/j.bbamem.2021.183772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/16/2021] [Accepted: 09/03/2021] [Indexed: 12/22/2022]
Abstract
NMOSD is a rare but severe relapsing remitting demyelinating disease that affects both adults and children. Most patients have pathogenic antibodies that target the central nervous system AQP4 protein. This review provides an update on our current understanding of the disease pathophysiology and describes the clinical, paraclinical features and therapeutic management of the disease.
Collapse
Affiliation(s)
- Sukhvir K Wright
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK; Dept. of Paediatric Neurology, The Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK.
| | - Evangeline Wassmer
- Institute of Health and Neurodevelopment, College of Health and Life Sciences, Aston University, Birmingham, UK; Dept. of Paediatric Neurology, The Birmingham Women's and Children's Hospital NHS Foundation Trust, Birmingham, UK
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| |
Collapse
|
14
|
Xiang W, Xie C, Luo J, Zhang W, Zhao X, Yang H, Cai Y, Ding J, Wang Y, Hao Y, Zhang Y, Guan Y. Low Frequency Ultrasound With Injection of NMO-IgG and Complement Produces Lesions Different From Experimental Autoimmune Encephalomyelitis Mice. Front Immunol 2021; 12:727750. [PMID: 34721390 PMCID: PMC8551829 DOI: 10.3389/fimmu.2021.727750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/20/2021] [Indexed: 11/23/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD), a relapsing autoimmune disease of the central nervous system, mainly targets the optic nerve and spinal cord. To date, all attempts at the establishment of NMOSD animal models have been based on neuromyelitis optica immunoglobulin G antibody (NMO-IgG) and mimic the disease in part. To solve this problem, we developed a rodent model by opening the blood-brain barrier (BBB) with low frequency ultrasound, followed by injection of NMO-IgG from NMOSD patients and complement to mice suffering pre-existing neuroinflammation produced by experimental autoimmune encephalomyelitis (EAE). In this study, we showed that ultrasound with NMO-IgG and complement caused marked inflammation and demyelination of both spinal cords and optic nerves compared to blank control group, as well as glial fibrillary acidic protein (GFAP) and aquaporin-4 (AQP4) loss of spinal cords and optic nerves compared to EAE mice and EAE mice with only BBB opening. In addition, magnetic resonance imaging (MRI) revealed optic neuritis with spinal cord lesions. We further demonstrated eye segregation defects in the dorsal lateral geniculate nucleus (dLGN) of these NMOSD mice.
Collapse
Affiliation(s)
- Weiwei Xiang
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chong Xie
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaying Luo
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xinxin Zhao
- Department of Radiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Yang
- Department of Neurology, The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, Shanghai, China
| | - Yu Cai
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Ding
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yishu Wang
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Hao
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Zhang
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Treatment of Neuromyelitis Optica Spectrum Disorders. Int J Mol Sci 2021; 22:ijms22168638. [PMID: 34445343 PMCID: PMC8395403 DOI: 10.3390/ijms22168638] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 12/11/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune central nervous system (CNS) inflammatory disorder that can lead to serious disability and mortality. Females are predominantly affected, including those within the reproductive age. Most patients develop relapsing attacks of optic neuritis; longitudinally extensive transverse myelitis; and encephalitis, especially brainstem encephalitis. The majority of NMOSD patients are seropositive for IgG autoantibodies against the water channel protein aquaporin-4 (AQP4-IgG), reflecting underlying aquaporin-4 autoimmunity. Histological findings of the affected CNS tissues of patients from in-vitro and in-vivo studies support that AQP4-IgG is directly pathogenic in NMOSD. It is believed that the binding of AQP4-IgG to CNS aquaporin-4 (abundantly expressed at the endfoot processes of astrocytes) triggers astrocytopathy and neuroinflammation, resulting in acute attacks. These attacks of neuroinflammation can lead to pathologies, including aquaporin-4 loss, astrocytic activation, injury and loss, glutamate excitotoxicity, microglial activation, neuroinflammation, demyelination, and neuronal injury, via both complement-dependent and complement-independent pathophysiological mechanisms. With the increased understanding of these mechanisms underlying this serious autoimmune astrocytopathy, effective treatments for both active attacks and long-term immunosuppression to prevent relapses in NMOSD are increasingly available based on the evidence from retrospective observational data and prospective clinical trials. Knowledge on the indications and potential side effects of these medications are essential for a clear evaluation of the potential benefits and risks to NMOSD patients in a personalized manner. Special issues such as pregnancy and the coexistence of other autoimmune diseases require additional concern and meticulous care. Future directions include the identification of clinically useful biomarkers for the prediction of relapse and monitoring of the therapeutic response, as well as the development of effective medications with minimal side effects, especially opportunistic infections complicated by long-term immunosuppression.
Collapse
|
16
|
Chen T, Bosco DB, Ying Y, Tian DS, Wu LJ. The Emerging Role of Microglia in Neuromyelitis Optica. Front Immunol 2021; 12:616301. [PMID: 33679755 PMCID: PMC7933531 DOI: 10.3389/fimmu.2021.616301] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Neuromyelitis optica (NMO) is an autoantibody-triggered neuro-inflammatory disease which preferentially attacks the spinal cord and optic nerve. Its defining autoantibody is specific for the water channel protein, aquaporin-4 (AQP4), which primarily is localized at the end-feet of astrocytes. Histopathology studies of early NMO lesions demonstrated prominent activation of microglia, the resident immune sentinels of the central nervous system (CNS). Significant microglial reactivity is also observed in NMO animal models induced by introducing AQP4-IgG into the CNS. Here we review the potential roles for microglial activation in human NMO patients as well as different animal models of NMO. We will focus primarily on the molecular mechanisms underlying microglial function and microglia-astrocyte interaction in NMO pathogenesis. Understanding the role of microglia in NMO pathology may yield novel therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Dai-Shi Tian
- Department of Neurology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
17
|
Richard C, Ruiz A, Cavagna S, Bigotte M, Vukusic S, Masaki K, Suenaga T, Kira JI, Giraudon P, Marignier R. Connexins in neuromyelitis optica: a link between astrocytopathy and demyelination. Brain 2021; 143:2721-2732. [PMID: 32889550 DOI: 10.1093/brain/awaa227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 01/26/2023] Open
Abstract
Neuromyelitis optica, a rare neuroinflammatory demyelinating disease of the CNS, is characterized by the presence of specific pathogenic autoantibodies directed against the astrocytic water channel aquaporin 4 (AQP4) and is now considered as an astrocytopathy associated either with complement-dependent astrocyte death or with astrocyte dysfunction. However, the link between astrocyte dysfunction and demyelination remains unclear. We propose glial intercellular communication, supported by connexin hemichannels and gap junctions, to be involved in demyelination process in neuromyelitis optica. Using mature myelinated cultures, we demonstrate that a treatment of 1 h to 48 h with immunoglobulins purified from patients with neuromyelitis optica (NMO-IgG) is responsible for a complement independent demyelination, compared to healthy donors' immunoglobulins (P < 0.001). In parallel, patients' immunoglobulins induce an alteration of connexin expression characterized by a rapid loss of astrocytic connexins at the membrane followed by an increased size of gap junction plaques (+60%; P < 0.01). This was co-observed with connexin dysfunction with gap junction disruption (-57%; P < 0.001) and increased hemichannel opening (+17%; P < 0.001), associated with glutamate release. Blocking connexin 43 hemichannels with a specific peptide was able to prevent demyelination in co-treatment with patients compared to healthy donors' immunoglobulins. By contrast, the blockade of connexin 43 gap junctions with another peptide was detrimental for myelin (myelin density -48%; P < 0.001). Overall, our results suggest that dysregulation of connexins would play a pathogenetic role in neuromyelitis optica. The further identification of mechanisms leading to connexin dysfunction and soluble factors implicated, would provide interesting therapeutic strategies for demyelinating disorders.
Collapse
Affiliation(s)
- Chloé Richard
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Anne Ruiz
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Sylvie Cavagna
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Maxime Bigotte
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Sandra Vukusic
- Service de neurologie, sclérose en plaques, pathologies de la myéline et neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France.,Centre de référence des maladies inflammatoires rares du cerveau et de la moelle, Lyon, France
| | - Katsuhisa Masaki
- Department of Neurology, Neurological institute, Graduate School of Medical Sciences, Kyushu University
| | | | - Jun-Ichi Kira
- Department of Neurology, Neurological institute, Graduate School of Medical Sciences, Kyushu University
| | - Pascale Giraudon
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Romain Marignier
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France.,Service de neurologie, sclérose en plaques, pathologies de la myéline et neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France.,Centre de référence des maladies inflammatoires rares du cerveau et de la moelle, Lyon, France
| |
Collapse
|
18
|
Luo J, Xie C, Zhang W, Cai Y, Ding J, Wang Y, Hao Y, Zhang Y, Guan Y. Experimental mouse model of NMOSD produced by facilitated brain delivery of NMO-IgG by microbubble-enhanced low-frequency ultrasound in experimental allergic encephalomyelitis mice. Mult Scler Relat Disord 2020; 46:102473. [PMID: 32919181 DOI: 10.1016/j.msard.2020.102473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/19/2020] [Accepted: 08/29/2020] [Indexed: 12/28/2022]
Abstract
Although optic neuritis and myelitis are the core clinical characteristics of neuromyelitis optica spectrum disorders (NMOSD), appropriate animal models of NMOSD with myelitis and optic neuritis are lacking. we developed a mouse model of NMOSD by intravenously injecting 100 µg neuromyelitis optica immunoglobulin G antibody (NMO-IgG) and complement into experimental allergic encephalomyelitis (EAE) mice after reversible blood-brain barrier (BBB) opening by microbubble-enhanced low-frequency ultrasound (MELFUS). Animals were assessed by histopathology. We found noticeable inflammation and demyelination concomitant with the loss of aquaporin-4 (AQP4) and glial fibrillary acidic protein (GFAP) expression in the spinal cord, brain and optic nerve, as well as human IgG and C9neo deposition. Thus, with the help of MELFUS, we established an NMOSD mouse model with the core lesions of NMOSD by applying a considerably lower dose of human NMO-IgG, which may help identify the pathogenesis and facilitate the development of other neuroimmune disease models in the future.
Collapse
Affiliation(s)
- Jiaying Luo
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Chong Xie
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Wei Zhang
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, 200233, Shanghai, China
| | - Yu Cai
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Jie Ding
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Yishu Wang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Yong Hao
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Ying Zhang
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China
| | - Yangtai Guan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No.1630 Dongfang Road, 200127, Shanghai, China.
| |
Collapse
|
19
|
Cobo-Calvo A, Ruiz A, Richard C, Blondel S, Cavagna S, Strazielle N, Ghersi-Egea JF, Giraudon P, Marignier R. Purified IgG from aquaporin-4 neuromyelitis optica spectrum disorder patients alters blood-brain barrier permeability. PLoS One 2020; 15:e0238301. [PMID: 32881954 PMCID: PMC7470361 DOI: 10.1371/journal.pone.0238301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neuromyelitis optica spectrum disorders (NMOSD) is a primary astrocytopathy driven by antibodies directed against the aquaporin-4 water channel located at the end-feet of the astrocyte. Although blood-brain barrier (BBB) breakdown is considered one of the key steps for the development and lesion formation, little is known about the molecular mechanisms involved. The aim of the study was to evaluate the effect of human immunoglobulins from NMOSD patients (NMO-IgG) on BBB properties. METHODS Freshly isolated brain microvessels (IBMs) from rat brains were used as a study model. At first, analysis of the secretome profile from IBMs exposed to purified NMO-IgG, to healthy donor IgG (Control-IgG), or non-treated, was performed. Second, tight junction (TJ) proteins expression in fresh IBMs and primary cultures of brain microvascular endothelial cells (BMEC) was analysed by Western blotting (Wb) after exposition to NMO-IgG and Control-IgG. Finally, functional BBB properties were investigated evaluating the presence of rat-IgG in tissue lysate from brain using Wb in the rat-model, and the passage of NMO-IgG and sucrose in a bicameral model. RESULTS We found that NMO-IgG induces functional and morphological BBB changes, including: 1) increase of pro-inflammatory cytokines production (CXCL-10 [IP-10], IL-6, IL-1RA, IL-1β and CXCL-3) in IBMs when exposed to NMO-IgG; 2) decrease of Claudin-5 levels by 25.6% after treatment of fresh IBMs by NMO-IgG compared to Control-IgG (p = 0.002), and similarly, decrease of Claudin-5 by at least 20% when BMEC were cultured with NMO-IgG from five different patients; 3) a higher level of rat-IgG accumulated in periventricular regions of NMO-rats compared to Control-rats and an increase in the permeability of BBB after NMO-IgG treatment in the bicameral model. CONCLUSION Human NMO-IgG induces both structural and functional alterations of BBB properties, suggesting a direct role of NMO-IgG on modulation of BBB permeability in NMOSD.
Collapse
Affiliation(s)
- Alvaro Cobo-Calvo
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuroinflammation and Centre de Référence Pour les Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM)–Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail:
| | - Anne Ruiz
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Chloé Richard
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Sandrine Blondel
- Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Department of Neurology/Neuroimmunology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sylvie Cavagna
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Nathalie Strazielle
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- BIP Facility, CRNL, Lyon, France
| | - Jean-François Ghersi-Egea
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
- BIP Facility, CRNL, Lyon, France
| | - Pascale Giraudon
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| | - Romain Marignier
- Service de Neurologie, Sclérose en Plaques, Pathologies de la Myéline et Neuroinflammation and Centre de Référence Pour les Maladies Inflammatoires Rares du Cerveau et de la Moelle (MIRCEM)–Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Neurosciences de Lyon, U1028 INSERM-CNRS UMR5292-UCBL, Bron, France
| |
Collapse
|
20
|
Yick LW, Tang CH, Ma OKF, Kwan JSC, Chan KH. Memantine ameliorates motor impairments and pathologies in a mouse model of neuromyelitis optica spectrum disorders. J Neuroinflammation 2020; 17:236. [PMID: 32782018 PMCID: PMC7418436 DOI: 10.1186/s12974-020-01913-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Neuromyelitis optica spectrum disorders (NMOSD) are central nervous system (CNS) autoimmune inflammatory demyelinating diseases characterized by recurrent episodes of acute optic neuritis and transverse myelitis. Aquaporin-4 immunoglobulin G (AQP4-IgG) autoantibodies, which target the water channel aquaporin-4 (AQP4) on astrocytic membrane, are pathogenic in NMOSD. Glutamate excitotoxicity, which is triggered by internalization of AQP4-glutamate transporter complex after AQP4-IgG binding to astrocytes, is involved in early NMOSD pathophysiologies. We studied the effects of memantine, a N-methyl-D-aspartate (NMDA) receptor antagonist, on motor impairments and spinal cord pathologies in mice which received human AQP4-IgG. Methods Purified IgG from AQP4-IgG-seropositive NMOSD patients were passively transferred to adult C57BL/6 mice with disrupted blood-brain barrier. Memantine was administered by oral gavage. Motor impairments of the mice were assessed by beam walking test. Spinal cords of the mice were assessed by immunofluorescence and ELISA. Results Oral administration of memantine ameliorated the motor impairments induced by AQP4-IgG, no matter the treatment was initiated before (preventive) or after (therapeutic) disease flare. Memantine profoundly reduced AQP4 and astrocyte loss, and attenuated demyelination and axonal loss in the spinal cord of mice which had received AQP4-IgG. The protective effects of memantine were associated with inhibition of apoptosis and suppression of neuroinflammation, with decrease in microglia activation and neutrophil infiltration and reduction of increase in levels of proinflammatory cytokines including interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). In addition, memantine elevated growth factors including brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), and vascular endothelial growth factor (VEGF) in the spinal cord. Conclusions Our findings support that glutamate excitotoxicity and neuroinflammation play important roles in complement-independent pathophysiology during early development of NMOSD lesions, and highlight the potential of oral memantine as a therapeutic agent in NMOSD acute attacks.
Collapse
Affiliation(s)
- Leung-Wah Yick
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Chi-Ho Tang
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Oscar Ka-Fai Ma
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jason Shing-Cheong Kwan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Koon-Ho Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong. .,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong. .,Department of Medicine, The University of Hong Kong, 4/F, Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, Hong Kong.
| |
Collapse
|
21
|
Ciappelloni S, Bouchet D, Dubourdieu N, Boué-Grabot E, Kellermayer B, Manso C, Marignier R, Oliet SHR, Tourdias T, Groc L. Aquaporin-4 Surface Trafficking Regulates Astrocytic Process Motility and Synaptic Activity in Health and Autoimmune Disease. Cell Rep 2020; 27:3860-3872.e4. [PMID: 31242419 DOI: 10.1016/j.celrep.2019.05.097] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 03/08/2019] [Accepted: 05/23/2019] [Indexed: 01/21/2023] Open
Abstract
Astrocytes constantly adapt their ramified morphology in order to support brain cell assemblies. Such plasticity is partly mediated by ion and water fluxes, which rely on the water channel aquaporin-4 (AQP4). The mechanism by which this channel locally contributes to process dynamics has remained elusive. Using a combination of single-molecule and calcium imaging approaches, we here investigated in hippocampal astrocytes the dynamic distribution of the AQP4 isoforms M1 and M23. Surface AQP4-M1 formed small aggregates that contrast with the large AQP4-M23 clusters that are enriched near glutamatergic synapses. Strikingly, stabilizing surface AQP4-M23 tuned the motility of astrocyte processes and favors glutamate synapse activity. Furthermore, human autoantibodies directed against AQP4 from neuromyelitis optica (NMO) patients impaired AQP4-M23 dynamic distribution and, consequently, astrocyte process and synaptic activity. Collectively, it emerges that the membrane dynamics of AQP4 isoform regulate brain cell assemblies in health and autoimmune brain disease targeting AQP4.
Collapse
Affiliation(s)
- Silvia Ciappelloni
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Delphine Bouchet
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Nadège Dubourdieu
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Eric Boué-Grabot
- Université de Bordeaux, 33077 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Blanka Kellermayer
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Constance Manso
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Romain Marignier
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Stéphane H R Oliet
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Thomas Tourdias
- Université de Bordeaux, 33077 Bordeaux, France; INSERM U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Laurent Groc
- Interdisciplinary Institute for NeuroSciences, CNRS UMR 5297, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France.
| |
Collapse
|
22
|
da Silva APB, Silva RBM, Goi LDS, Molina RD, Machado DC, Sato DK. Experimental Models of Neuroimmunological Disorders: A Review. Front Neurol 2020; 11:389. [PMID: 32477252 PMCID: PMC7235321 DOI: 10.3389/fneur.2020.00389] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022] Open
Abstract
Immune-mediated inflammatory diseases of the central nervous system (CNS) are a group of neurological disorders in which inflammation and/or demyelination are induced by cellular and humoral immune responses specific to CNS antigens. They include diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSD), acute disseminated encephalomyelitis (ADEM) and anti-NMDA receptor encephalitis (NMDAR encephalitis). Over the years, many in vivo and in vitro models were used to study clinical, pathological, physiological and immunological features of these neuroimmunological disorders. Nevertheless, there are important aspects of human diseases that are not fully reproduced in the experimental models due to their technical limitations. In this review, we describe the preclinical models of neuroimmune disorders, and how they contributed to the understanding of these disorders and explore potential treatments. We also describe the purpose and limitation of each one, as well as the recent advances in this field.
Collapse
Affiliation(s)
- Ana Paula Bornes da Silva
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Rodrigo Braccini Madeira Silva
- Research Center in Toxicology and Pharmacology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Leise Daniele Sckenal Goi
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Rachel Dias Molina
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Denise Cantarelli Machado
- School of Medicine, Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,Molecular and Cellular Biology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Douglas Kazutoshi Sato
- Neuroinflammation and Neuroimmunology Laboratory, Brain Institute, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Graduate Program in Pediatrics and Child Health, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.,School of Medicine, Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
23
|
Assessing the anterior visual pathway in optic neuritis: recent experimental and clinical aspects. Curr Opin Neurol 2020; 32:346-357. [PMID: 30694926 DOI: 10.1097/wco.0000000000000675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) and related autoimmune disorders of the central nervous system such as neuromyelitis optica spectrum disorders (NMOSD) are characterized by chronic disability resulting from autoimmune neuroinflammation, with demyelination, astrocyte damage, impaired axonal transmission and neuroaxonal loss. Novel therapeutics stopping or reversing the progression of disability are still urgently warranted. This review addresses research on optic neuritis in preclinical experimental models and their translation to clinical trials. RECENT FINDINGS Optic neuritis can be used as paradigm for an MS relapse which can serve to evaluate the efficacy of novel therapeutics in clinical trials with a reasonable duration and cohort size. The advantage is the linear structure of the visual pathway allowing the assessment of visual function and retinal structure as highly sensitive outcome parameters. Experimental autoimmune encephalomyelitis is an inducible, inflammatory and demyelinating central nervous system disease extensively used as animal model of MS. Optic neuritis is part of the clinicopathological manifestations in a number of experimental autoimmune encephalomyelitis models. These have gained increasing interest for studies evaluating neuroprotective and/or remyelinating substances as longitudinal, visual and retinal readouts have become available. SUMMARY Translation of preclinical experiments, evaluating neuroprotective or remyelinating therapeutics to clinical studies is challenging. In-vivo readouts like optical coherence tomography, offers the possibility to transfer experimental study designs to clinical optic neuritis trials.
Collapse
|
24
|
Faissner S, Graz F, Reinehr S, Petrikowski L, Haupeltshofer S, Ceylan U, Stute G, Winklmeier S, Pache F, Paul F, Ruprecht K, Meinl E, Dick HB, Gold R, Kleiter I, Joachim SC. Binding patterns and functional properties of human antibodies to AQP4 and MOG on murine optic nerve and retina. J Neuroimmunol 2020; 342:577194. [PMID: 32143071 DOI: 10.1016/j.jneuroim.2020.577194] [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: 12/17/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 11/19/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune-inflammatory CNS disease affecting spinal cord and optic nerves, mediated by autoantibodies against aquaporin-4 (AQP4) and myelin-oligodendrocyte-glycoprotein (MOG). Effects of those immunoglobulins (Ig) on retina and optic nerve are incompletely understood. We investigated AQP4-IgG and MOG-IgG sera on retina and optic nerve ex vivo and in 2D2 mice, which harbor a transgenic MOG-specific T-cell receptor. Some sera reacted with murine retina and optic nerve showing distinct binding patterns, suggesting different epitopes being targeted in both subgroups. Transfer of total IgG from a MOG-IgG positive patient to 2D2 mice did neither enhance disability nor induce functional or histological alterations in the retina.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany.
| | - Florian Graz
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany; Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Sabrina Reinehr
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Laura Petrikowski
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany; Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Steffen Haupeltshofer
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany
| | - Ulaş Ceylan
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany
| | - Gesa Stute
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Stephan Winklmeier
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Florence Pache
- Department of Neurology, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Friedemann Paul
- Department of Neurology, Charité - Universitaetsmedizin Berlin, Berlin, Germany; NeuroCure Clinical Research Center und Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany
| | - Ingo Kleiter
- Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany; Marianne-Strauß-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke, Berg, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892 Bochum, Germany; Department of Neurology, Ruhr-University Bochum, St. Josef-Hospital, Gudrunstr. 56, 44791 Bochum, Germany.
| |
Collapse
|
25
|
Jarius S, Wildemann B. The history of neuromyelitis optica. Part 2: 'Spinal amaurosis', or how it all began. J Neuroinflammation 2019; 16:280. [PMID: 31883522 PMCID: PMC6935230 DOI: 10.1186/s12974-019-1594-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/23/2019] [Indexed: 01/08/2023] Open
Abstract
Neuromyelitis optica (NMO) was long considered a clinical variant of multiple sclerosis (MS). However, the discovery of a novel and pathogenic anti-astrocytic serum autoantibody targeting aquaporin-4 (termed NMO-IgG or AQP4-Ab), the most abundant water channel protein in the central nervous system, led to the recognition of NMO as a distinct disease entity in its own right and generated strong and persisting interest in the condition. NMO is now studied as a prototypic autoimmune disorder, which differs from MS in terms of immunopathogenesis, clinicoradiological presentation, optimum treatment, and prognosis. While the history of classic MS has been extensively studied, relatively little is known about the history of NMO. In Part 1 of this series we focused on the late 19th century, when the term 'neuromyelitis optica' was first coined, traced the term's origins and followed its meandering evolution throughout the 20th and into the 21st century. Here, in Part 2, we demonstrate that the peculiar concurrence of acute optic nerve and spinal cord affliction characteristic for NMO caught the attention of physicians much earlier than previously thought by re-presenting a number of very early cases of possible NMO that date back to the late 18th and early 19th century. In addition, we comprehensively discuss the pioneering concept of 'spinal amaurosis', which was introduced into the medical literature by ophthalmologists in the first half of the 19th century.
Collapse
Affiliation(s)
- S. Jarius
- Department of Neurology, Molecular Neuroimmunology Group, University of Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| | - B. Wildemann
- Department of Neurology, Molecular Neuroimmunology Group, University of Heidelberg, Otto Meyerhof Center, Im Neuenheimer Feld 350, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Yao X, Adams MS, Jones PD, Diederich CJ, Verkman AS. Noninvasive, Targeted Creation of Neuromyelitis Optica Pathology in AQP4-IgG Seropositive Rats by Pulsed Focused Ultrasound. J Neuropathol Exp Neurol 2019; 78:47-56. [PMID: 30500945 DOI: 10.1093/jnen/nly107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (herein called NMO) is an autoimmune disease of the CNS characterized by astrocyte injury, inflammation, and demyelination. In seropositive NMO, immunoglobulin G autoantibodies against aquaporin-4 (AQP4-IgG) cause primary astrocyte injury. A passive transfer model of NMO was developed in which spatially targeted access of AQP4-IgG into the CNS of seropositive rats was accomplished by pulsed focused ultrasound through intact skin. Following intravenous administration of microbubbles, pulsed ultrasound at 0.5 MPa peak acoustic pressure was applied using a 1 MHz transducer with 6-cm focal length. In brain, the transient opening of the blood-brain barrier (BBB) in an approximately prolate ellipsoidal volume of diameter ∼3.5 mm and length ∼44 mm allowed entry of IgG-size molecules for up to 3-6 hours. The ultrasound treatment did not cause erythrocyte extravasation or inflammation. Ultrasound treatment in AQP4-IgG seropositive rats produced localized NMO pathology in brain, with characteristic astrocyte injury, inflammation, and demyelination after 5 days. Pathology was not seen when complement was inhibited, when non-NMO human IgG was administered instead of AQP4-IgG, or in AQP4-IgG seropositive AQP4 knockout rats. NMO pathology was similarly created in cervical spinal cord in seropositive rats. These results establish a noninvasive, spatially targeted model of NMO in rats, and demonstrate that BBB permeabilization, without underlying injury or inflammation, is sufficient to create NMO pathology in AQP4-IgG seropositive rats.
Collapse
Affiliation(s)
| | - Matthew S Adams
- Department of Medicine and Physiology.,Thermal Therapy Research Group, Department of Radiation Oncology, University of California, San Francisco, California
| | - Peter D Jones
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California, San Francisco, California
| | - Chris J Diederich
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California, San Francisco, California
| | | |
Collapse
|
27
|
Duan T, Verkman AS. Experimental animal models of aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders: progress and shortcomings. Brain Pathol 2019; 30:13-25. [PMID: 31587392 DOI: 10.1111/bpa.12793] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) is a heterogeneous group of neuroinflammatory conditions associated with demyelination primarily in spinal cord and optic nerve, and to a lesser extent in brain. Most NMOSD patients are seropositive for IgG autoantibodies against aquaporin-4 (AQP4-IgG), the principal water channel in astrocytes. There has been interest in establishing experimental animal models of seropositive NMOSD (herein referred to as NMO) in order to elucidate NMO pathogenesis mechanisms and to evaluate drug candidates. An important outcome of early NMO animal models was evidence for a pathogenic role of AQP4-IgG. However, available animal models of NMO, based largely on passive transfer to rodents of AQP4-IgG or transfer of AQP4-sensitized T cells, often together with pro-inflammatory maneuvers, only partially recapitulate the clinical and pathological features of human NMO, and are inherently biased toward humoral or cellular immune mechanisms. This review summarizes current progress and shortcomings in experimental animal models of seropositive NMOSD, and opines on the import of advancing animal models.
Collapse
Affiliation(s)
- Tianjiao Duan
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143.,Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143
| |
Collapse
|
28
|
Wu Y, Zhong L, Geng J. Visual impairment in neuromyelitis optica spectrum disorders (NMOSD). J Chem Neuroanat 2019; 97:66-70. [DOI: 10.1016/j.jchemneu.2019.01.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/31/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022]
|
29
|
Repetitive intrathecal injection of human NMO-IgG with complement exacerbates disease severity with NMO pathology in experimental allergic encephalomyelitis mice. Mult Scler Relat Disord 2019; 30:225-230. [PMID: 30825702 DOI: 10.1016/j.msard.2019.02.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 02/16/2019] [Accepted: 02/23/2019] [Indexed: 12/31/2022]
Abstract
Neuromyelitis optica (NMO) is recognized as a different CNS autoimmune disease from multiple sclerosis (MS). Whether NMO-IgG contributes directly to the pathogenesis of NMO or is just a serologic marker of autoimmune responses of the disease needs to be clarified. We created MOG-induced experimental autoimmune encephalomyelitis (EAE) mice by passively transferring NMO-IgG to model the pathogenic findings in NMO patients. The mice were divided into three groups and administered intrathecal PBS, human complement with IgG from normal subjects, or IgG from AQP4(+) patients on days 8 and 11 after immunization. The EAE scores of EAE mice with intrathecal NMO-IgG injection were significantly elevated 14 days post-immunization. All of the mice were sacrificed for brain and spinal cord pathology analysis on day 21 post-immunization. Compared to mice given normal human IgG, EAE mice injected with NMO-IgG had markedly decreased AQP4 and glial fibrillary acidic protein (GFAP) expression and fluorescent intensity in the brain and spinal cord but more scattered deposition of complement (C9neo). Thus, our studies not only support the pathogenic role of NMO-IgG with complement in NMO disease but also provide a platform for the development of future therapeutics.
Collapse
|
30
|
Cobo-Calvo A, Ayrignac X, Kerschen P, Horellou P, Cotton F, Labauge P, Vukusic S, Deiva K, Serguera C, Marignier R. Cranial nerve involvement in patients with MOG antibody-associated disease. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e543. [PMID: 30800725 PMCID: PMC6384017 DOI: 10.1212/nxi.0000000000000543] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/07/2018] [Indexed: 12/16/2022]
Abstract
Objective To describe clinical and radiologic features of cranial nerve (CN) involvement in patients with myelin oligodendrocyte glycoprotein antibodies (MOG-IgG) and to assess the potential underlying mechanism of CN involvement using a nonhuman primate (NHP) model. Methods Epidemiologic, clinical, and radiologic features from a national cohort of 273 MOG-IgG–positive patients were retrospectively reviewed for CN involvement between January 2014 and January 2018. MOG-IgG binding was evaluated in CNS, CN, and peripheral nerve tissues from NHP. Results We identified 3 MOG-IgG–positive patients with radiologic and/or clinical CN involvement. Two patients displayed either trigeminal or vestibulocochlear nerve lesions at the root level, and the remaining patient had an oculomotor nerve involvement at the root exit and at the cisternal level. Additional CNS involvement was found in all 3 patients. None of the 3 patients' sera recognized MOG expression in CN of NHP. Conclusion Craneal nerve involvement can coexist in patients with MOG antibody disease, although the underlying pathophysiology remains elusive.
Collapse
Affiliation(s)
- Alvaro Cobo-Calvo
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Xavier Ayrignac
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Philippe Kerschen
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Philippe Horellou
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Francois Cotton
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Pierre Labauge
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Sandra Vukusic
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Kumaran Deiva
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Ché Serguera
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| | - Romain Marignier
- Service de neurologie (A.C.-C., S.V., R.M.), sclérose en plaques, pathologies de la myéline et neuroinflammation and Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle (MIRCEM)- Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, France; Lyon Neuroscience Research Center (A.C.-C., R.M.), U1028 INSERM, UMR5292 CNRS, FLUID Team, Lyon, France; Service de sclérose en plaques (X.A., P.L.), Hôpital Universitaire de Montpellier, France; Service de Neurologie (P.K.), Centre hospitalier de Luxembourg; Inserm (P.H.), U 1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud 11, CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Faculté de médecine, Le Kremlin-Bicêtre Cedex, France; Service de Radiologie (F.C.), Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France; Université Claude Bernard Lyon 1 (F.C., S.V., R.M.), F-69100 Villeurbanne, France; Lyon's Neuroscience Research Center (S.V.), Observatoire Français de la Sclérose en Plaques, INSERM 1028 et CNRS UMR5292, Lyon, France; Service de neurologie pédiatrique (K.D.), Centre de référence pour les maladies inflammatoires rares du cerveau et de la moelle, Le Kremlin-Bicêtre, France; and INSERM US27 MIRCen (C.S.), CEA, Fontenay-aux-Roses, France
| |
Collapse
|
31
|
Wu Y, Zhong L, Geng J. Neuromyelitis optica spectrum disorder: Pathogenesis, treatment, and experimental models. Mult Scler Relat Disord 2018; 27:412-418. [PMID: 30530071 DOI: 10.1016/j.msard.2018.12.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/21/2018] [Accepted: 12/02/2018] [Indexed: 01/10/2023]
Abstract
Neuromyelitis optica (NMO) and NMO spectrum disorder (NMOSD) are inflammatory CNS syndromes mainly involving the optic nerve and/or spinal cord and characterized by the presence of serum aquaporin-4 immunoglobulin G antibodies (AQP4-IgG). The pathology of NMOSD is complicated, while therapies for NMOSD are limited and only partially effective in most cases. This review article focuses on the main pathology of NMOSD involving AQP4-IgG and lymphocyte function. We also review the existing therapeutic methods and potential new treatments. Experimental NMO animal models are crucial for further research into NMO pathology and treatment. However, no AQP4-IgG-immunized animals have been reported. The establishment of NMO models is therefore difficult and primarily depends on the generation of transgenic mice or transcranial manipulation using human or monoclonal mouse anti-AQP4 antibodies. Advantages and disadvantages of each model are discussed.
Collapse
Key Words
- APC, antigen-presenting cell
- Abbreviations: ADCC, antibody-dependent cellular cytotoxicity
- Aqp4, aquaporin 4
- Aquaporin-4
- BAFF, b-cell activating factor
- BBB, blood-brain barrier
- BCR, b cell receptor
- CDD, complement-dependent cytotoxicity
- CFA, complete freund's adjuvant
- CSF, cerebrospinal fluid
- CXCL, c-x-c motif chemokine ligand
- EAE, experimental autoimmune encephalomyelitis
- ECD, extracellular domain
- Experimental animal models
- IGG, immunoglobulin g
- IVMP, methylprednisolone pulse
- LETM, longitudinally extensive transverse myelitis
- MAB, monoclonal antibody
- MBP, myelin-binding protein
- MOG, myelin oligodendrocyte glycoprotein
- MOG-Ab, anti-MOG antibody
- NF-H, neurofilament heavy chain
- NMO, neuromyelitis optica
- NMO-IgG, NMO with serum AQP4-IgG
- NMOSD, NMO spectrum disorder
- Neuromyelitis optica
- Neuromyelitis optica spectrum disorder
- PB, plasmablast
- PP, plasmapheresis
- Remyelination
Collapse
Affiliation(s)
- Yan Wu
- Department of Neurology, Xichang Road No.295, Kunming 650000, China.
| | - Lianmei Zhong
- Department of Neurology, Xichang Road No.295, Kunming 650000, China
| | - Jia Geng
- Department of Neurology, Xichang Road No.295, Kunming 650000, China
| |
Collapse
|
32
|
|
33
|
Zhang Y, Bao Y, Qiu W, Peng L, Fang L, Xu Y, Yang H. Structural and visual functional deficits in a rat model of neuromyelitis optica spectrum disorders related optic neuritis. Exp Eye Res 2018; 175:124-132. [DOI: 10.1016/j.exer.2018.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/30/2018] [Accepted: 06/13/2018] [Indexed: 12/15/2022]
|
34
|
Yick LW, Ma OKF, Ng RCL, Kwan JSC, Chan KH. Aquaporin-4 Autoantibodies From Neuromyelitis Optica Spectrum Disorder Patients Induce Complement-Independent Immunopathologies in Mice. Front Immunol 2018; 9:1438. [PMID: 29988553 PMCID: PMC6026644 DOI: 10.3389/fimmu.2018.01438] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/11/2018] [Indexed: 11/13/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are central nervous system inflammatory disorders causing significant morbidities and mortality. The majority of NMOSD patients have autoimmunity against aquaporin-4 (AQP4), evidenced by seropositivity for autoantibodies against aquaporin-4 (AQP4–IgG). AQP4–IgG is pathogenic with neuroinflammation initiated upon binding of AQP4–IgG to astrocytic AQP4. Complement activation contributes to astrocytic cytotoxicity, neuroinflammation, and tissue necrosis in NMOSD, but the role of complement-independent mechanisms is uncertain. We studied the complement-independent pathogenic effects of AQP4–IgG by passive transfer of IgG from NMOSD patients to mice with breached blood–brain barrier (BBB). Mice, pretreated with bacterial proteins, received daily intraperitoneal injections of IgG purified from AQP4–IgG-seropositive NMOSD patients [IgG(AQP4+)], or IgG from AQP4–IgG-seronegative patients [IgG(AQP4−)] or healthy subjects [IgG(Healthy)] for 8 days. Motor function was tested by walking across narrow beams, and spinal cords were collected for immunofluorescent analysis. We found that human IgG infiltrated into cord parenchyma of mice with breached BBB without deposition of complement activation products. Spinal cord of mice that received IgG(AQP4+) demonstrated loss of AQP4 and glial fibrillary acidic protein (suggestive of astrocyte loss), decrease in excitatory amino acid transporter 2, microglial/macrophage activation, neutrophil infiltration, patchy demyelination, and loss in axonal integrity. Mice that received IgG(AQP4+) required longer time with more paw slips to walk across narrow beams indicative of motor slowing and incoordination. Our findings suggest that AQP4–IgG induces complement-independent cord pathologies, including astrocytopathy, neuroinflammation, demyelination, and axonal injuries/loss, which are associated with subtle motor impairments. These complement-independent pathophysiologies likely contribute to early NMOSD lesion development.
Collapse
Affiliation(s)
- Leung-Wah Yick
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Oscar Ka-Fai Ma
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Roy Chun-Laam Ng
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jason Shing-Cheong Kwan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Koon-Ho Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Neuroimmunology and Neuroinflammation Research Laboratory, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
35
|
NMO-IgG and AQP4 Peptide Can Induce Aggravation of EAMG and Immune-Mediated Muscle Weakness. J Immunol Res 2018; 2018:5389282. [PMID: 29951558 PMCID: PMC5987235 DOI: 10.1155/2018/5389282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/20/2018] [Accepted: 04/02/2018] [Indexed: 12/31/2022] Open
Abstract
Neuromyelitis optica (NMO) and myasthenia gravis (MG) are autoimmune diseases mediated by autoantibodies against either aquaporin 4 (AQP4) or acetylcholine receptor (AChR), respectively. Recently, we and others have reported an increased prevalence of NMO in patients with MG. To verify whether coexisting autoimmune disease may exacerbate experimental autoimmune MG, we tested whether active immunization with AQP4 peptides or passive transfer of NMO-Ig can affect the severity of EAMG. Injection of either AQP4 peptide or NMO-Ig to EAMG or to naive mice caused increased fatigability and aggravation of EAMG symptoms as expressed by augmented muscle weakness (but not paralysis), decremental response to repetitive nerve stimulation, increased neuromuscular jitter, and aberration of immune responses. Thus, our study shows increased disease severity in EAMG mice following immunization with the NMO autoantigen AQP4 or by NMO-Ig, mediated by augmented inflammatory response. This can explain exacerbation or increased susceptibility of patients with one autoimmune disease to develop additional autoimmune syndrome.
Collapse
|
36
|
Mørch MT, Sørensen SF, Khorooshi R, Asgari N, Owens T. Selective localization of IgG from cerebrospinal fluid to brain parenchyma. J Neuroinflammation 2018; 15:110. [PMID: 29665816 PMCID: PMC5904996 DOI: 10.1186/s12974-018-1159-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/11/2018] [Indexed: 11/29/2022] Open
Abstract
Background Encounter of autoantibodies with specific antigens can lead to hypersensitivity reactions and pathology. In multiple sclerosis and neuromyelitis optica spectrum disease (NMOSD), immunoglobulin-G (IgG) deposition has been observed in pathological lesions in the central nervous system. The paradigmatic autoantibodies in NMOSD are specific for the water channel aquaporin-4, localized to astrocytic end-feet at the blood-brain barrier and ependymal cells at the cerebrospinal fluid-brain barrier. We have previously observed that IgG antibodies from NMO patients (NMO-IgG) access brain parenchyma from the cerebrospinal fluid and induce subpial and periventricular NMO-like lesions and blood-brain barrier breakdown, in a complement-dependent manner. Objective To investigate how IgG trafficking from cerebrospinal fluid to brain parenchyma can be influenced by injury. Methods IgG from healthy donors was intrathecally injected into the cerebrospinal fluid via cisterna magna at 1, 2, 4, or 7 days after a distal stereotactic sterile needle insertion to the striatum. Results Antibody deposition, detected by staining for human IgG, peaked 1 day after the intrathecal injection and was selectively seen close to the needle insertion. When NMO-IgG was intrathecally injected, we observed complement-dependent NMO-like pathology (loss of aquaporin-4 and glial fibrillary acidic protein) proximal to the insertion site, with similar kinetics. A fluorescent tracer did not show the same distribution indicating IgG-selective localization. Conclusion These findings suggest that IgG from cerebrospinal fluid localize selectively in brain parenchyma at the site of injury and pathogenic NMO-IgG induce astrocyte pathology at the same location.
Collapse
Affiliation(s)
- Marlene Thorsen Mørch
- Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, JB. Winsloewsvej 25, 5000, Odense, Denmark
| | - Sofie Forsberg Sørensen
- Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, JB. Winsloewsvej 25, 5000, Odense, Denmark
| | - Reza Khorooshi
- Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, JB. Winsloewsvej 25, 5000, Odense, Denmark
| | - Nasrin Asgari
- Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, JB. Winsloewsvej 25, 5000, Odense, Denmark.,Department of Neurology, Slagelse Hospital, Institute of Regional Health Research, Slagelse, Denmark
| | - Trevor Owens
- Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, JB. Winsloewsvej 25, 5000, Odense, Denmark.
| |
Collapse
|
37
|
Combination Treatment of C16 Peptide and Angiopoietin-1 Alleviates Neuromyelitis Optica in an Experimental Model. Mediators Inflamm 2018; 2018:4187347. [PMID: 29670463 PMCID: PMC5835265 DOI: 10.1155/2018/4187347] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/22/2017] [Accepted: 12/05/2017] [Indexed: 12/25/2022] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune inflammatory demyelinating disease that mainly affects the spinal cord and optic nerve, causing blindness and paralysis in some individuals. Moreover, NMO may cause secondary complement-dependent cytotoxicity (CDC), leading to oligodendrocyte and neuronal damage. In this study, a rodent NMO model, showing typical NMO pathogenesis, was induced with NMO-IgG from patient serum and human complement. We then tested whether the combination of C16, an αvβ3 integrin-binding peptide, and angiopoietin-1 (Ang1), a member of the endothelial growth factor family, could alleviate NMO in the model. Our results demonstrated that this combination therapy significantly decreased disease severity, inflammatory cell infiltration, secondary demyelination, and axonal loss, thus reducing neural death. In conclusion, our study suggests a possible treatment that can relieve progressive blindness and paralysis in an animal model of NMO through improvement of the inflammatory milieu.
Collapse
|
38
|
Meli R, Pirozzi C, Pelagalli A. New Perspectives on the Potential Role of Aquaporins (AQPs) in the Physiology of Inflammation. Front Physiol 2018; 9:101. [PMID: 29503618 PMCID: PMC5820367 DOI: 10.3389/fphys.2018.00101] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022] Open
Abstract
Aquaporins (AQPs) are emerging, in the last few decades, as critical proteins regulating water fluid homeostasis in cells involved in inflammation. AQPs represent a family of ubiquitous membrane channels that regulate osmotically water flux in various tissues and sometimes the transport of small solutes, including glycerol. Extensive data indicate that AQPs, working as water channel proteins, regulate not only cell migration, but also common events essential for inflammatory response. The involvement of AQPs in several inflammatory processes, as demonstrated by their dysregulation both in human and animal diseases, identifies their new role in protection and response to different noxious stimuli, including bacterial infection. This contribution could represent a new key to clarify the dilemma of host-pathogen communications, and opens up new scenarios regarding the investigation of the modulation of specific AQPs, as target for new pharmacological therapies. This review provides updated information on the underlying mechanisms of AQPs in the regulation of inflammatory responses in mammals and discusses the broad spectrum of options that can be tailored for different diseases and their pharmacological treatment.
Collapse
Affiliation(s)
- Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
- Institute of Biostructure and Bioimaging, National Research Council (CNR), Naples, Italy
| |
Collapse
|
39
|
Srisupa - Olan T, Siritho S, Kittisares K, Jitprapaikulsan J, Sathukitchai C, Prayoonwiwat N. Beneficial effect of plasma exchange in acute attack of neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2018; 20:115-121. [DOI: 10.1016/j.msard.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 12/30/2017] [Accepted: 01/12/2018] [Indexed: 12/16/2022]
|
40
|
Marignier R, Cobo Calvo A, Vukusic S. Neuromyelitis optica and neuromyelitis optica spectrum disorders. Curr Opin Neurol 2018; 30:208-215. [PMID: 28306572 DOI: 10.1097/wco.0000000000000455] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The discovery of highly specific auto-antibodies directed against the water channel aquaporin 4 was a quantum leap in the definition, classification and management of neuromyelitis optica (NMO). Herein, we propose an update on epidemiological, clinical and therapeutic advances in the field, underlining unmet needs. RECENT FINDINGS Large-scale epidemiological studies have recently provided a more precise evaluation of NMO prevalence and a better stratification regarding ethnicity and sex. New criteria have been proposed for so-called NMO spectrum disorders (NMOSD) and their relevance is currently being assessed. The identification of a new clinical entity associated to antibodies against myelin oligodendrocyte glycoprotein questions the border of NMOSD. SUMMARY The concept of NMOSD is opening a new era in clinical practice, allowing an easier and more homogeneous diagnosis and an increase in newly identified cases. This will facilitate clinical studies and support new therapeutic trial. Future researches should focus on the position of seronegative NMOSD and myelin oligodendrocyte glycoprotein-IgG disorders in the field and on promising strategies, including the immune tolerisation approaches, to eventually cure NMO.
Collapse
Affiliation(s)
- Romain Marignier
- aSclérose en plaques, pathologies de la myéline et neuro-inflammation, Hospices Civils de Lyon bCentre de Recherche en Neurosciences de Lyon, Inserm U1028 CNRS UMR5292, FLUID team, Faculté de Médecine Laennec cObservatoire Français de la Sclérose en Plaques (OFSEP), Lyon, France
| | | | | |
Collapse
|
41
|
Yao X, Verkman AS. Marked central nervous system pathology in CD59 knockout rats following passive transfer of Neuromyelitis optica immunoglobulin G. Acta Neuropathol Commun 2017; 5:15. [PMID: 28212662 PMCID: PMC5316191 DOI: 10.1186/s40478-017-0417-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 02/03/2017] [Indexed: 11/10/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (herein called NMO) is an inflammatory demyelinating disease of the central nervous system in which pathogenesis involves complement-dependent cytotoxicity (CDC) produced by immunoglobulin G autoantibodies targeting aquaporin-4 (AQP4-IgG) on astrocytes. We reported evidence previously, using CD59-/- mice, that the membrane-associated complement inhibitor CD59 modulates CDC in NMO (Zhang and Verkman, J. Autoimmun. 53:67-77, 2014). Motivated by the observation that rats, unlike mice, have human-like complement activity, here we generated CD59-/- rats to investigate the role of CD59 in NMO and to create NMO pathology by passive transfer of AQP4-IgG under conditions in which minimal pathology is produced in normal rats. CD59-/- rats generated by CRISPR/Cas9 technology showed no overt phenotype at baseline except for mild hemolysis. CDC assays in astrocyte cultures and cerebellar slices from CD59-/- rats showed much greater sensitivity to AQP4-IgG and complement than those from CD59+/+ rats. Intracerebral administration of AQP4-IgG in CD59-/- rats produced marked NMO pathology, with astrocytopathy, inflammation, deposition of activated complement, and demyelination, whereas identically treated CD59+/+ rats showed minimal pathology. A single, intracisternal injection of AQP4-IgG in CD59-/- rats produced hindlimb paralysis by 3 days, with inflammation and deposition of activated complement in spinal cord, optic nerves and brain periventricular and surface matter, with most marked astrocyte injury in cervical spinal cord. These results implicate an important role of CD59 in modulating NMO pathology in rats and demonstrate amplification of AQP4-IgG-induced NMO disease with CD59 knockout.
Collapse
|
42
|
Stiebel-Kalish H, Lotan I, Brody J, Chodick G, Bialer O, Marignier R, Bach M, Hellmann MA. Retinal Nerve Fiber Layer May Be Better Preserved in MOG-IgG versus AQP4-IgG Optic Neuritis: A Cohort Study. PLoS One 2017; 12:e0170847. [PMID: 28125740 PMCID: PMC5268377 DOI: 10.1371/journal.pone.0170847] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/11/2017] [Indexed: 12/21/2022] Open
Abstract
Background Optic neuritis (ON) in patients with anti-myelin oligodendrocyte glycoprotein (MOG)-IgG antibodies has been associated with a better clinical outcome than anti-aquaporin 4 (AQP4)- IgG ON. Average retinal nerve fiber layer thickness (RNFL) correlates with visual outcome after ON. Objectives The aim of this study was to examine whether anti-MOG-IgG ON is associated with better average RNFL compared to anti-AQP4-IgG ON, and whether this corresponds with a better visual outcome. Methods A retrospective study was done in a consecutive cohort of patients following anti-AQP4-IgG and anti-MOG-IgG ON. A generalized estimating equation (GEE) models analysis was used to compare average RNFL outcomes in ON eyes of patients with MOG-IgG to AQP4-IgG-positive patients, after adjusting for the number of ON events. The final mean visual field defect and visual acuity were compared between ON eyes of MOG-IgG and AQP4-IgG-positive patients. A correlation between average RNFL and visual function was performed in all study eyes. Results Sixteen patients were analyzed; ten AQP4-IgG-positive and six MOG-IgG-positive. The six patients with MOG-IgG had ten ON events with disc edema, five of which were bilateral. In the AQP4-IgG-positive ON events, 1/10 patients had disc edema. Final average RNFL was significantly better in eyes following MOG-IgG-ON (75.33μm), compared to 63.63μm in AQP4-IgG-ON, after adjusting for the number of ON attacks (GEE, p = 0.023). Mean visual field defects were significantly smaller (GEE, p = 0.046) among MOG-IgG positive ON eyes compared to AQP-IgG positive ON eyes, but last visual acuity did not differ between the groups (GEE, p = 0.153). Among all eyes, average RNFL positively correlated with mean visual field defect (GEE, p = 0.00015) and negatively correlated with final visual acuity (GEE, p = 0.00005). Conclusions Following ON, RNFL is better preserved in eyes of patients with MOG-IgG antibodies compared to those with AQP4-IgG antibodies, correlating with better visual outcomes.
Collapse
Affiliation(s)
- Hadas Stiebel-Kalish
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuro-Ophthalmology Unit, Department of Ophthalmology, Rabin Medical Center, Petah Tikva, Israel
- * E-mail:
| | - Itay Lotan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuro-Immunology Service and Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
| | - Judith Brody
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuro-Ophthalmology Unit, Department of Ophthalmology, Rabin Medical Center, Petah Tikva, Israel
| | - Gabriel Chodick
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Epidemiology and Preventive Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omer Bialer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuro-Ophthalmology Unit, Department of Ophthalmology, Rabin Medical Center, Petah Tikva, Israel
| | - Romain Marignier
- Service de Neurologie A, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, Lyon-Bron, France
| | - Michael Bach
- Eye Center, Medical Center, University of Freiburg, Germany, and Faculty of Medicine, University of Freiburg, Germany
| | - Mark Andrew Hellmann
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neuro-Immunology Service and Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
43
|
Felix CM, Levin MH, Verkman AS. Complement-independent retinal pathology produced by intravitreal injection of neuromyelitis optica immunoglobulin G. J Neuroinflammation 2016; 13:275. [PMID: 27765056 PMCID: PMC5072328 DOI: 10.1186/s12974-016-0746-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/13/2016] [Indexed: 12/19/2022] Open
Abstract
Background Neuromyelitis optica (NMO), an autoimmune inflammatory disease of the central nervous system, is often associated with retinal abnormalities including thinning of the retinal nerve fiber layer and microcystic changes. Here, we demonstrate that passive transfer of an anti-aquaporin-4 autoantibody (AQP4-IgG) produces primary retinal pathology. Methods AQP4-IgG was delivered to adult rat retinas by intravitreal injection. Rat retinas and retinal explant cultures were assessed by immunofluorescence. Results Immunofluorescence showed AQP4-IgG deposition on retinal Müller cells, with greatly reduced AQP4 expression and increased glial fibrillary acidic protein by 5 days. There was mild retinal inflammation with microglial activation but little leukocyte infiltration and loss of retinal ganglion cells by 30 days with thinning of the ganglion cell complex. Interestingly, the loss of AQP4 was complement independent as seen in cobra venom factor-treated rats and in normal rats administered a mutated AQP4-IgG lacking complement effector function. Exposure of ex vivo retinal cultures to AQP4-IgG produced a marked reduction in AQP4 expression by 24 h, which was largely prevented by inhibitors of endocytosis or lysosomal acidification. Conclusions Passive transfer of AQP4-IgG results in primary, complement-independent retinal pathology, which might contribute to retinal abnormalities seen in NMO patients.
Collapse
Affiliation(s)
- Christian M Felix
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.,Departments of Medicine and Physiology, University of California, San Francisco, 1246 Health Sciences East Tower, San Francisco, CA, 94143-0521, USA
| | - Marc H Levin
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA.,Department of Ophthalmology, The Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, 1246 Health Sciences East Tower, San Francisco, CA, 94143-0521, USA.
| |
Collapse
|