1
|
Bradl M, Yu Q, Takai Y. The immunological processes behind aquaporin 4-antibody seropositive neuromyelitis optica spectrum disorders. Semin Immunol 2025; 78:101945. [PMID: 40154151 DOI: 10.1016/j.smim.2025.101945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Ever since the discovery of pathogenic aquaporin 4-specific antibodies in the serum of patients with neuromyelitis optica spectrum disorders current knowledge about clinical observations and diagnosis, and about the underlying pathology and resulting therapies have been put forward in excellent reviews and primary publications. However, in order to further develop novel strategies for the treatment of this disease, there is an urgent need to understand the immunological processes associated with the formation of the pathogenic antibodies, and with aberrant immune responses observed in affected patients. In this review, we will highlight and evaluate important studies on these processes.
Collapse
Affiliation(s)
- Monika Bradl
- Medical University Vienna, Center for Brain Research, Division of Neuroimmunology, Austria.
| | - Qian Yu
- Medical University Vienna, Center for Brain Research, Division of Neuroimmunology, Austria
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Pathology, Tohoku University Hospital, Sendai, Japan
| |
Collapse
|
2
|
Afzali AM, Ulianov O, Eckardt L, Stas I, Seeholzer L, Steiger K, Merkler D, Korn T. AQP4-specific T cells determine lesion localization in the CNS in a model of NMOSD. Acta Neuropathol Commun 2025; 13:27. [PMID: 39934927 PMCID: PMC11817536 DOI: 10.1186/s40478-025-01947-8] [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: 10/02/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a paradigmatic autoimmune disease of the central nervous system (CNS), in which the water channel protein Aquaporin-4 (AQP4) is targeted by a self-reactive immune response. While the immunopathology of human NMOSD is largely dependent on antibodies to astrocytic AQP4, the role of AQP4-specific T cells for the localization and quality of NMOSD lesions in the CNS is not known. Only recently, we established that thymic B cells express and present AQP4 in the context of MHC class II molecules to purge the naive T cell receptor repertoire of AQP4-specific clones. Here, we exploited this finding to investigate the lesion localization in the CNS of B cell conditional AQP4-deficient (Aqp4ΔB) mice, which harbor AQP4-specific precursors in their naive T cell repertoire and can be sensitized to mount a strong AQP4(201-220)-specific CD4+ T cell response. Sensitization of Aqp4ΔB mice with AQP4(201-220) was sufficient to induce clinical disease. The spatiotemporal lesion distribution and the glial cell response in AQP4(201-220)-induced experimental autoimmune encephalomyelitis (EAE) was compared to classical MOG(35-55)-induced EAE in Aqp4ΔB mice. In contrast to MOG-EAE, AQP4(201-220)-induced EAE was characterized by midline lesions in the brain, retinal pathology, and lesions at the grey matter/white matter border zone in the spinal cord. Therefore, we conclude that antigen-specific T cells dictate the localization of NMOSD-lesions in the CNS.
Collapse
Affiliation(s)
- Ali Maisam Afzali
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany
| | - Oleksii Ulianov
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany
| | - Luise Eckardt
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany
| | - Ingrid Stas
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany
| | - Lea Seeholzer
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, Centre Médical Universitaire, 1, Rue Michel Servet, 1211, Geneva, Switzerland
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Ismaninger Str. 22, 81675, Munich, Germany.
- Department of Neurology, Technical University of Munich School of Medicine, Ismaninger Str. 22, 81675, Munich, Germany.
- Munich Cluster for Systems Neurology, Feodor-Lynen-Str. 17, 81377, Munich, Germany.
| |
Collapse
|
3
|
Pressley KR, Schwegman L, De Oca Arena MM, Huizar CC, Zamvil SS, Forsthuber TG. HLA-transgenic mouse models to study autoimmune central nervous system diseases. Autoimmunity 2024; 57:2387414. [PMID: 39167553 PMCID: PMC11470778 DOI: 10.1080/08916934.2024.2387414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/23/2024]
Abstract
It is known that certain human leukocyte antigen (HLA) genes are associated with autoimmune central nervous system (CNS) diseases, such as multiple sclerosis (MS), but their exact role in disease susceptibility and etiopathogenesis remains unclear. The best studied HLA-associated autoimmune CNS disease is MS, and thus will be the primary focus of this review. Other HLA-associated autoimmune CNS diseases, such as autoimmune encephalitis and neuromyelitis optica will be discussed. The lack of animal models to accurately capture the complex human autoimmune response remains a major challenge. HLA transgenic (tg) mice provide researchers with powerful tools to investigate the underlying mechanisms promoting susceptibility and progression of HLA-associated autoimmune CNS diseases, as well as for elucidating the myelin epitopes potentially targeted by T cells in autoimmune disease patients. We will discuss the potential role(s) of autoimmune disease-associated HLA alleles in autoimmune CNS diseases and highlight information provided by studies using HLA tg mice to investigate the underlying pathological mechanisms and opportunities to use these models for development of novel therapies.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Neuroscience, Developmental, and Regenerative Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Lance Schwegman
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | | | - Carol Chase Huizar
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Scott S. Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
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
|
5
|
Sánchez A, Padilla Y, Lorduy A, Sanchez J, Munera M, Baena C, Bernal C, Urrego J. In silico analysis of molecular mimicry between human aquaporin 3, Aspergillus fumigatus aquaporin and aquaporins from allergic sources. F1000Res 2024; 13:358. [PMID: 39310813 PMCID: PMC11415755 DOI: 10.12688/f1000research.142843.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition that has a significant impact on quality of life. The immune response and allergy symptoms in AD are triggered by the recognition of specific allergens by IgE antibodies. Cross-reactivity can lead to auto-IgE responses, potentially worsening AD symptoms. Our research aimed to enhance our understanding of allergenic sources, including A. fumigatus, and their role in AD. We focused on molecular mimicry between human AQP3 and A. fumigatus aquaporin. Methods In our in-silico analysis, we compared the amino acid sequences of human aquaporin 3 (AQP3) and A. fumigatus aquaporin with 25 aquaporins from various allergenic sources, sourced from the UniProt and NCBI databases. Phylogenetic relationship analysis and homology-based modeling were conducted. We identified conserved antigenic regions located within the 3D structures. Results The global identity levels among the studied aquaporins averaged 32.6%. One antigenic site exhibited a remarkable local region, with a conserved identity of 71.4%. We categorized the aquaporins into five monophyletic clades (A-E), with group B showing the highest identity (95%), including six mammalian aquaporins, including AQP3. When comparing A. fumigatus aquaporins, the highest identity was observed with Malassezia sympodialis at 35%. Both human and A. fumigatus aquaporins have three linear and three discontinuous epitopes. Conclusions We identified potential linear and conformational epitopes of AQP3, indicating a possible molecular mimicry between humans and A. fumigatus aquaporins. This suggests autoreactivity and potential cross-reactivity, although further validation using in vitro and in vivo experiments is required.
Collapse
Affiliation(s)
- Andrés Sánchez
- Group of Research Medicine (GINUMED), Rafael Nunez University Corporation, Cartagena, Bolívar, Colombia
- Clinical and Experimental Allergy Group (GACE), University of Antioquia, Medellín, Antioquia, Colombia
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| | - Yaquelin Padilla
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| | - Adriana Lorduy
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| | - Jorge Sanchez
- Clinical and Experimental Allergy Group (GACE), University of Antioquia, Medellín, Antioquia, Colombia
| | - Marlon Munera
- Group of Research Medicine (GINUMED), Rafael Nunez University Corporation, Cartagena, Bolívar, Colombia
| | - Claudia Baena
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| | - Carlos Bernal
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| | - Juan Urrego
- Research Group of Pharmaceutical, Cosmetic, and Food Technology (GITFCA), University of Cartagena, Cartagena, Bolívar, Colombia
| |
Collapse
|
6
|
Arellano G, Loda E, Chen Y, Neef T, Cogswell AC, Primer G, Joy G, Kaschke K, Wills S, Podojil JR, Popko B, Balabanov R, Miller SD. Interferon-γ controls aquaporin 4-specific Th17 and B cells in neuromyelitis optica spectrum disorder. Brain 2024; 147:1344-1361. [PMID: 37931066 PMCID: PMC10994540 DOI: 10.1093/brain/awad373] [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: 01/27/2023] [Revised: 09/27/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a CNS autoimmune inflammatory disease mediated by T helper 17 (Th17) and antibody responses to the water channel protein, aquaporin 4 (AQP4), and associated with astrocytopathy, demyelination and axonal loss. Knowledge about disease pathogenesis is limited and the search for new therapies impeded by the absence of a reliable animal model. In our work, we determined that NMOSD is characterized by decreased IFN-γ receptor signalling and that IFN-γ depletion in AQP4201-220-immunized C57BL/6 mice results in severe clinical disease resembling human NMOSD. Pathologically, the disease causes autoimmune astrocytic and CNS injury secondary to cellular and humoral inflammation. Immunologically, the absence of IFN-γ allows for increased expression of IL-6 in B cells and activation of Th17 cells, and generation of a robust autoimmune inflammatory response. Consistent with NMOSD, the experimental disease is exacerbated by administration of IFN-β, whereas repletion of IFN-γ, as well as therapeutic targeting of IL-17A, IL-6R and B cells, ameliorates it. We also demonstrate that immune tolerization with AQP4201-220-coupled poly(lactic-co-glycolic acid) nanoparticles could both prevent and effectively treat the disease. Our findings enhance the understanding of NMOSD pathogenesis and provide a platform for the development of immune tolerance-based therapies, avoiding the limitations of the current immunosuppressive therapies.
Collapse
Affiliation(s)
- Gabriel Arellano
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eileah Loda
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yanan Chen
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Tobias Neef
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew C Cogswell
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Grant Primer
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Godwin Joy
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Kevin Kaschke
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Samantha Wills
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, USA
| | - Joseph R Podojil
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- COUR Pharmaceutical Development Company, Inc., Northbrook, IL 60077, USA
| | - Brian Popko
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Roumen Balabanov
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Afzali AM, Nirschl L, Sie C, Pfaller M, Ulianov O, Hassler T, Federle C, Petrozziello E, Kalluri SR, Chen HH, Tyystjärvi S, Muschaweckh A, Lammens K, Delbridge C, Büttner A, Steiger K, Seyhan G, Ottersen OP, Öllinger R, Rad R, Jarosch S, Straub A, Mühlbauer A, Grassmann S, Hemmer B, Böttcher JP, Wagner I, Kreutzfeldt M, Merkler D, Pardàs IB, Schmidt Supprian M, Buchholz VR, Heink S, Busch DH, Klein L, Korn T. B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4. Nature 2024; 627:407-415. [PMID: 38383779 PMCID: PMC10937377 DOI: 10.1038/s41586-024-07079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.
Collapse
Affiliation(s)
- Ali Maisam Afzali
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Lucy Nirschl
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Christopher Sie
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Monika Pfaller
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Oleksii Ulianov
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Tobias Hassler
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Christine Federle
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Elisabetta Petrozziello
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Sudhakar Reddy Kalluri
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Hsin Hsiang Chen
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sofia Tyystjärvi
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Andreas Muschaweckh
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Katja Lammens
- Department of Biochemistry at the Gene Center, Ludwig-Maximilians-University, Munich, Germany
| | - Claire Delbridge
- Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Department of Neuropathology, Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Gönül Seyhan
- Institute for Experimental Hematology, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Ole Petter Ottersen
- Division of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Adrian Straub
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Anton Mühlbauer
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Simon Grassmann
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bernhard Hemmer
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Geneva Faculty of Medicine, Centre Médical Universitaire, Geneva, Switzerland
| | | | - Marc Schmidt Supprian
- Institute for Experimental Hematology, TranslaTUM Cancer Center, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Sylvia Heink
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich School of Medicine and Health, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Ludger Klein
- Biomedical Center (BMC), Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine and Health, Munich, Germany.
- Department of Neurology, Technical University of Munich School of Medicine and Health, Munich, Germany.
- Munich Cluster for Systems Neurology, Munich, Germany.
| |
Collapse
|
8
|
Jiang Y, Dai S, Pang R, Qin L, Zhang M, Liu H, Wang X, Zhang J, Peng G, Wang Y, Li W. Single-cell RNA sequencing reveals cell type-specific immune regulation associated with human neuromyelitis optica spectrum disorder. Front Immunol 2024; 15:1322125. [PMID: 38440735 PMCID: PMC10909925 DOI: 10.3389/fimmu.2024.1322125] [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: 10/15/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction One rare type of autoimmune disease is called neuromyelitis optica spectrum disorder (NMOSD) and the peripheral immune characteristics of NMOSD remain unclear. Methods Here, single-cell RNA sequencing (scRNA-seq) is used to characterize peripheral blood mononuclear cells from individuals with NMOSD. Results The differentiation and activation of lymphocytes, expansion of myeloid cells, and an excessive inflammatory response in innate immunity are observed. Flow cytometry analyses confirm a significant increase in the percentage of plasma cells among B cells in NMOSD. NMOSD patients exhibit an elevated percentage of CD8+ T cells within the T cell population. Oligoclonal expansions of B cell receptors are observed after therapy. Additionally, individuals with NMOSD exhibit elevated expression of CXCL8, IL7, IL18, TNFSF13, IFNG, and NLRP3. Discussion Peripheral immune response high-dimensional single-cell profiling identifies immune cell subsets specific to a certain disease and identifies possible new targets for NMOSD.
Collapse
Affiliation(s)
- Yushu Jiang
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuhua Dai
- Department of Neurology, Zhoukou Central Hospital, Zhoukou, Henan, China
| | - Rui Pang
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lingzhi Qin
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Milan Zhang
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huiqin Liu
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojuan Wang
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiewen Zhang
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Gongxin Peng
- Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yongchao Wang
- Department of Neurology, People’s Hospital of Yexian, Pingdingshan, Henan, China
| | - Wei Li
- Department of Neurology, Henan Joint International Research Laboratory Of Accurate Diagnosis, Treatment, Research And Development, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Aspden JW, Murphy MA, Kashlan RD, Xiong Y, Poznansky MC, Sîrbulescu RF. Intruders or protectors - the multifaceted role of B cells in CNS disorders. Front Cell Neurosci 2024; 17:1329823. [PMID: 38269112 PMCID: PMC10806081 DOI: 10.3389/fncel.2023.1329823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
B lymphocytes are immune cells studied predominantly in the context of peripheral humoral immune responses against pathogens. Evidence has been accumulating in recent years on the diversity of immunomodulatory functions that B cells undertake, with particular relevance for pathologies of the central nervous system (CNS). This review summarizes current knowledge on B cell populations, localization, infiltration mechanisms, and function in the CNS and associated tissues. Acute and chronic neurodegenerative pathologies are examined in order to explore the complex, and sometimes conflicting, effects that B cells can have in each context, with implications for disease progression and treatment outcomes. Additional factors such as aging modulate the proportions and function of B cell subpopulations over time and are also discussed in the context of neuroinflammatory response and disease susceptibility. A better understanding of the multifactorial role of B cell populations in the CNS may ultimately lead to innovative therapeutic strategies for a variety of neurological conditions.
Collapse
Affiliation(s)
- James W. Aspden
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Matthew A. Murphy
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rommi D. Kashlan
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Yueyue Xiong
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Ruxandra F. Sîrbulescu
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Remlinger J, Bagnoud M, Meli I, Massy M, Linington C, Chan A, Bennett JL, Hoepner R, Enzmann V, Salmen A. Modelling MOG antibody-associated disorder and neuromyelitis optica spectrum disorder in animal models: Spinal cord manifestations. Mult Scler Relat Disord 2023; 78:104892. [PMID: 37499337 PMCID: PMC11792092 DOI: 10.1016/j.msard.2023.104892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/18/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
Antibodies to myelin oligodendrocyte glycoprotein (MOG-IgG) or aquaporin 4 (AQP4-IgG) are associated with CNS inflammatory disorders. We directly compared MOG35-55-induced experimental autoimmune encephalomyelitis exacerbated by MOG- and AQP4-IgG (versus isotype IgG, Iso-IgG). Disease severity was highest after MOG-IgG application. MOG- and AQP4-IgG administration increased disease incidence compared to Iso-IgG. Inflammatory lesions appeared earlier and with distinct localizations after AQP4-IgG administration. AQP4 intensity was more reduced after AQP4- than MOG-IgG administration at acute disease phase. The described models are suitable for comparative analyses of pathological features associated with MOG- and AQP4-IgG and the investigation of therapeutic interventions.
Collapse
Affiliation(s)
- Jana Remlinger
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Maud Bagnoud
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Ivo Meli
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Marine Massy
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, 3010, Switzerland
| | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States of America
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Volker Enzmann
- Department of Ophthalmology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Bern, 3010, Switzerland.
| |
Collapse
|
11
|
Remlinger J, Bagnoud M, Meli I, Massy M, Hoepner R, Linington C, Chan A, Bennett JL, Enzmann V, Salmen A. Modeling MOG Antibody-Associated Disorder and Neuromyelitis Optica Spectrum Disorder in Animal Models: Visual System Manifestations. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200141. [PMID: 37429715 PMCID: PMC10691219 DOI: 10.1212/nxi.0000000000200141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND AND OBJECTIVES Mechanisms of visual impairment in aquaporin 4 antibody (AQP4-IgG) seropositive neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody (MOG-IgG)-associated disorder (MOGAD) are incompletely understood. The respective impact of optic nerve demyelination and primary and secondary retinal neurodegeneration are yet to be investigated in animal models. METHODS Active MOG35-55 experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6Jrj mice, and monoclonal MOG-IgG (8-18C5, murine), recombinant AQP4-IgG (rAb-53, human), or isotype-matched control IgG (Iso-IgG, human) was administered 10 days postimmunization. Mobility impairment was scored daily. Visual acuity by optomotor reflex and ganglion cell complex thickness (GCC, 3 innermost retinal layers) by optical coherence tomography (OCT) were longitudinally assessed. Histopathology of optic nerve and retina was investigated during presymptomatic, acute, and chronic disease phases for immune cells, demyelination, complement deposition, natural killer (NK) cell, AQP4, and astrocyte involvement, retinal ganglion cells (RGCs), and Müller cell activation. Groups were compared by nonparametric tests with a p value <0.05 indicating statistical significance. RESULTS Visual acuity decreased from baseline to chronic phase in MOG-IgG (mean ± standard error of the mean: 0.54 ± 0.01 to 0.46 ± 0.02 cycles/degree, p < 0.05) and AQP4-IgG EAE (0.54 ± 0.01 to 0.43 ± 0.02, cycles/degree, p < 0.05). Immune cell infiltration of optic nerves started in presymptomatic AQP4-IgG, but not in MOG-IgG EAE (5.85 ± 2.26 vs 0.13 ± 0.10 macrophages/region of interest [ROI] and 1.88 ± 0.63 vs 0.15 ± 0.06 T cells/ROI, both p < 0.05). Few NK cells, no complement deposition, and stable glial fibrillary acid protein and AQP4 fluorescence intensity characterized all EAE optic nerves. Lower GCC thickness (Spearman correlation coefficient r = -0.44, p < 0.05) and RGC counts (r = -0.47, p < 0.05) correlated with higher mobility impairment. RGCs decreased from presymptomatic to chronic disease phase in MOG-IgG (1,705 ± 51 vs 1,412 ± 45, p < 0.05) and AQP4-IgG EAE (1,758 ± 14 vs 1,526 ± 48, p < 0.01). Müller cell activation was not observed in either model. DISCUSSION In a multimodal longitudinal characterization of visual outcome in animal models of MOGAD and NMOSD, differential retinal injury and optic nerve involvement were not conclusively clarified. Yet optic nerve inflammation was earlier in AQP4-IgG-associated pathophysiology. Retinal atrophy determined by GCC thickness (OCT) and RGC counts correlating with mobility impairment in the chronic phase of MOG-IgG and AQP4-IgG EAE may serve as a generalizable marker of neurodegeneration.
Collapse
Affiliation(s)
- Jana Remlinger
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Maud Bagnoud
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Ivo Meli
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Marine Massy
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Robert Hoepner
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Christopher Linington
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Andrew Chan
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Jeffrey L Bennett
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Volker Enzmann
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland
| | - Anke Salmen
- From the Department of Neurology (J.R., M.B., I.M., M.M., R.H., A.C., A.S.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences (J.R., M.M.), University of Bern, Switzerland; Institute of Infection (C.L.), Immunity and Inflammation, University of Glasgow, UK; Departments of Neurology and Ophthalmology (J.L.B.), Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Aurora; and Department of Ophthalmology (V.E.), Inselspital, Bern University Hospital and Department for BioMedical Research (DBMR), University of Bern, Switzerland.
| |
Collapse
|
12
|
Sagan SA, Moinfar Z, Moseley CE, Dandekar R, Spencer CM, Verkman AS, Ottersen OP, Sobel RA, Sidney J, Sette A, Anderson MS, Steinman L, Wilson MR, Sabatino JJ, Zamvil SS. T cell deletional tolerance restricts AQP4 but not MOG CNS autoimmunity. Proc Natl Acad Sci U S A 2023; 120:e2306572120. [PMID: 37463205 PMCID: PMC10372680 DOI: 10.1073/pnas.2306572120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Aquaporin-4 (AQP4)-specific Th17 cells are thought to have a central role in neuromyelitis optica (NMO) pathogenesis. When modeling NMO, only AQP4-reactive Th17 cells from AQP4-deficient (AQP4-/-), but not wild-type (WT) mice, caused CNS autoimmunity in recipient WT mice, indicating that a tightly regulated mechanism normally ensures tolerance to AQP4. Here, we found that pathogenic AQP4 T cell epitopes bind MHC II with exceptionally high affinity. Examination of T cell receptor (TCR) α/β usage revealed that AQP4-specific T cells from AQP4-/- mice employed a distinct TCR repertoire and exhibited clonal expansion. Selective thymic AQP4 deficiency did not fully restore AQP4-reactive T cells, demonstrating that thymic negative selection alone did not account for AQP4-specific tolerance in WT mice. Indeed, AQP4-specific Th17 cells caused paralysis in recipient WT or B cell-deficient mice, which was followed by complete recovery that was associated with apoptosis of donor T cells. However, donor AQP4-reactive T cells survived and caused persistent paralysis in recipient mice deficient in both T and B cells or mice lacking T cells only. Thus, AQP4 CNS autoimmunity was limited by T cell-dependent deletion of AQP4-reactive T cells. In contrast, myelin oligodendrocyte glycoprotein (MOG)-specific T cells survived and caused sustained disease in WT mice. These findings underscore the importance of peripheral T cell deletional tolerance to AQP4, which may be relevant to understanding the balance of AQP4-reactive T cells in health and in NMO. T cell tolerance to AQP4, expressed in multiple tissues, is distinct from tolerance to MOG, an autoantigen restricted in its expression.
Collapse
Affiliation(s)
- Sharon A Sagan
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Zahra Moinfar
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Carson E Moseley
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Ravi Dandekar
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Collin M Spencer
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| | - Alan S Verkman
- Department of Medicine, University of California, San Francisco, CA 94143
- Department of Physiology, University of California, San Francisco, CA 94143
| | - Ole Petter Ottersen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo NO-0316, Norway
| | - Raymond A Sobel
- Department of Pathology, Stanford University School of Medicine, Palo Alto VA Health Care System, Palo Alto, CA 94305
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Mark S Anderson
- Program in Immunology, University of California, San Francisco, CA 94143
- Diabetes Center, University of California, San Francisco, CA 94143
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305
| | - Michael R Wilson
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Joseph J Sabatino
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA 94143
- Program in Immunology, University of California, San Francisco, CA 94143
| |
Collapse
|
13
|
Serizawa K, Miyake S, Katsura Y, Yorozu K, Kurasawa M, Tomizawa-Shinohara H, Yasuno H, Matsumoto Y. Intradermal AQP4 peptide immunization induces clinical features of neuromyelitis optica spectrum disorder in mice. J Neuroimmunol 2023; 380:578109. [PMID: 37210799 DOI: 10.1016/j.jneuroim.2023.578109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
We challenged to create a mouse model of neuromyelitis optica spectrum disorder (NMOSD) induced by AQP4 peptide immunization. Intradermal immunization with AQP4 p201-220 peptide induced paralysis in C57BL/6J mice, but not in AQP4 KO mice. AQP4 peptide-immunized mice showed pathological features similar to NMOSD. Administration of anti-IL-6 receptor antibody (MR16-1) inhibited the induction of clinical signs and prevented the loss of GFAP/AQP4 and deposition of complement factors in AQP4 peptide-immunized mice. This novel experimental model may contribute to further understanding the pathogenesis of NMOSD, elucidating the mechanism of action of therapeutic agents, and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Kenichi Serizawa
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan.
| | - Shota Miyake
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Yoshichika Katsura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Keigo Yorozu
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Mitsue Kurasawa
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | | | - Hideyuki Yasuno
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Yoshihiro Matsumoto
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| |
Collapse
|
14
|
Nicosia M, Lee J, Beavers A, Kish D, Farr GW, McGuirk PR, Pelletier MF, Lathia JD, Fairchild RL, Valujskikh A. Water channel aquaporin 4 is required for T cell receptor mediated lymphocyte activation. J Leukoc Biol 2023; 113:544-554. [PMID: 36805947 PMCID: PMC10848298 DOI: 10.1093/jleuko/qiad010] [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: 01/09/2022] [Revised: 12/16/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Aquaporins are a family of ubiquitously expressed transmembrane water channels implicated in a broad range of physiological functions. We have previously reported that aquaporin 4 (AQP4) is expressed on T cells and that treatment with a small molecule AQP4 inhibitor significantly delays T cell mediated heart allograft rejection. Using either genetic deletion or small molecule inhibitor, we show that AQP4 supports T cell receptor mediated activation of both mouse and human T cells. Intact AQP4 is required for optimal T cell receptor (TCR)-related signaling events, including nuclear translocation of transcription factors and phosphorylation of proximal TCR signaling molecules. AQP4 deficiency or inhibition impairs actin cytoskeleton rearrangements following TCR crosslinking, causing inferior TCR polarization and a loss of TCR signaling. Our findings reveal a novel function of AQP4 in T lymphocytes and identify AQP4 as a potential therapeutic target for preventing TCR-mediated T cell activation.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Ashley Beavers
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Danielle Kish
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - George W. Farr
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Paul R. McGuirk
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Marc F. Pelletier
- Aeromics Inc., 470 James Street Suite 007, New Haven, CT 06513, United States
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Robert L. Fairchild
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| |
Collapse
|
15
|
Nishiyama S, Wright AE, Lotan I, Mikami T, Paul F, Aoki M, Levy M. Upregulated complement receptors correlate with Fc gamma receptor 3A-positive natural killer and natural killer-T cells in neuromyelitis optica spectrum disorder. J Neuroinflammation 2022; 19:296. [PMID: 36503481 PMCID: PMC9743562 DOI: 10.1186/s12974-022-02661-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Inhibition of terminal complement in neuromyelitis optica spectrum disorder (NMOSD) using eculizumab helps prevent relapses, but the exact mechanism of action of the drug remains unclear. Similarly, genetic variants in the Fc Gamma receptor 3A (FCGR3A), also known as CD16, are correlated with outcomes in NMOSD, but the immune cells expressing those CD16 are unknown. We compared CD16 expression on immune cells modulated by complement activity in natural killer (NK) cells and natural killer-T (NKT) cells in NMOSD to disease and normal-healthy controls. METHODS Peripheral blood cell (PBMC) samples from 45 patients with NMOSD with aquaporin 4 (AQP4)-IgG, 18 disease controls, and 19 normal controls were analyzed for CD16 expression and complement receptors in vitro. RESULTS At baseline, the number of NKT cells was increased in NMOSD (p < 0.001), but the proportion that was CD16 positive was lower compared to normal and disease controls (p = 0.0012). NK cell count was normal, but the ratio that was CD16 positive was also significantly lower (p < 0.001). In both NK cells and NKT cells from NMOSD, C5 complement receptor expression was much higher than normal and disease controls (p < 0.001 for both). We also evaluated activation markers CD69 and CD83, which were also significantly higher in NK and NKT cells from NMOSD patients. FCGR3A p158 V/V genotype group in NMOSD patients showed decreased NK cell proportion with activation, and fewer CD16-expressing NKT cells than the F/F genotype group. DISCUSSION Our results support an immunopathogenesis model in which complement pathway activation in NK/NKT cells upregulates CD16 expression that binds to antibody/antigen complexes. In the context of NMOSD, these complement-sensitive cells may be responsible for the escalating autoimmune activity.
Collapse
Affiliation(s)
- Shuhei Nishiyama
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Building 114, 16th St., Room 3150, Charlestown, MA 02129 Boston, USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA ,grid.69566.3a0000 0001 2248 6943Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi Japan
| | - Amy E. Wright
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Building 114, 16th St., Room 3150, Charlestown, MA 02129 Boston, USA
| | - Itay Lotan
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Building 114, 16th St., Room 3150, Charlestown, MA 02129 Boston, USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Takahisa Mikami
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Building 114, 16th St., Room 3150, Charlestown, MA 02129 Boston, USA ,grid.67033.310000 0000 8934 4045Department of Neurology, Tufts University School of Medicine, Boston, MA USA
| | - Friedemann Paul
- grid.6363.00000 0001 2218 4662Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Masashi Aoki
- grid.69566.3a0000 0001 2248 6943Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Miyagi Japan
| | - Michael Levy
- grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Building 114, 16th St., Room 3150, Charlestown, MA 02129 Boston, USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| |
Collapse
|
16
|
Mora Cuervo DL, Hansel G, Sato DK. Immunobiology of neuromyelitis optica spectrum disorders. Curr Opin Neurobiol 2022; 76:102618. [PMID: 35973380 DOI: 10.1016/j.conb.2022.102618] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 11/03/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a rare autoimmune inflammatory disease of the central nervous system. Most of the cases are positive for autoantibodies targeting the water channel aquaporin-4 (AQP4-IgG). Activated B and T cells, innate immunity cells, pro-inflammatory cytokines, and activated complement contribute to the formation of the NMOSD lesions. Optic neuritis, longitudinally extensive myelitis, and area postrema syndrome are core clinical manifestations. NMOSD diagnosis is based on clinical manifestations, magnetic resonance imaging findings, and AQP4-IgG positivity. Cell-based assays are the preferred method for the detection of AQP4-IgG. Acute relapses are treated with IV methylprednisolone or plasma exchange. Recent advances on the NMOSD immunobiology led to approved treatments such as eculizumab, satralizumab, and inebilizumab.
Collapse
Affiliation(s)
- Daissy Liliana Mora Cuervo
- Medicine and Health Sciences Postgraduation Program, School of Medicine, Pontificial Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil. https://twitter.com/DaissyMora
| | - Gisele Hansel
- Medicine and Health Sciences Postgraduation Program, School of Medicine, Pontificial Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil; Neuroinflammation and Neuroimmunology Lab, Brain Institute of Rio Grande Do Sul, Porto Alegre, Brazil
| | - Douglas Kazutoshi Sato
- Medicine and Health Sciences Postgraduation Program, School of Medicine, Pontificial Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil; Neuroinflammation and Neuroimmunology Lab, Brain Institute of Rio Grande Do Sul, Porto Alegre, Brazil; Institute of Geriatrics and Gerontology, Pontificial Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil.
| |
Collapse
|
17
|
Zhou Y, Song S, Han Y, Liu J, Yin B, Yuan C, Guo R, Jia Z, Sun Y, Li B. Altered non-coding RNA profiles and potential disease marker identification in peripheral blood mononuclear cells of patients with NMOSD. Int Immunopharmacol 2022; 109:108899. [PMID: 35689955 DOI: 10.1016/j.intimp.2022.108899] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/11/2022] [Accepted: 05/24/2022] [Indexed: 11/19/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelination disorder, and dysregulation of RNAs contributes to its pathogenesis. We aimed to reveal the expression profiles of RNAs, including messenger RNA (mRNA), circular RNA (circRNA) and long non-coding RNA (lncRNA), in the peripheral blood mononuclear cells (PBMCs) of patients with NMOSD. Seven NMOSD patients and seven healthy controls (HCs) were enrolled in the competitive endogenous RNA (ceRNA) microarray analysis. Bioinformatics analysis was then performed on the microarray data. Selected RNAs were validated by RT-qPCR. Differentially expressed (DE) RNA profiles of patients and HCs were related to NK cell mediated cytotoxicity, the IL-17 signaling pathway, and the B cell receptor signaling pathway. Moreover, DE non-coding RNAs (DE ncRNAs) including DE circRNAs and DE lncRNAs, may participate in the transforming growth factor beta (TGF-β) signaling pathway, leukocyte migration and neutrophil chemotaxis. Immune cell infiltration analysis showed that the abundance of M1 macrophages and plasma cells significantly increased, while that of M2 macrophages significantly decreased in the NMOSD group. Finally, through RT-qPCR validation, lnc-HELZ-7:1 (95% confidential interval of area under curve [95%CI of AUC] = 0.6633-1.0000), ring finger protein-LIM domain interacting (RLIM; 95%CI of AUC = 0.6980-1.0000), and hsa_circ_0026993 (95%CI of AUC = 0.7550-1.0000) could discriminate NMOSD from HCs by receiver operating characteristic curve analysis. To our knowledge, this is the first study to preliminarily investigate the RNA profiles, especially circRNA profiles in PBMCs of NMOSD patients from North China. We identified lnc-HELZ-7:1, RLIM, and hsa_circ_0026993 as the potential disease markers for NMOSD.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Shuang Song
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Yusen Han
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Jia Liu
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Bowen Yin
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Congcong Yuan
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Ruoyi Guo
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Zhen Jia
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Yafei Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China
| | - Bin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China; Key Laboratory of Neurology of Hebei Province, The Second Hospital of Hebei Medical University, City Shijiazhuang 050000, Province Hebei, PR China.
| |
Collapse
|
18
|
Vakrakou A, Chatzistamatiou T, Koros C, Karathanasis D, Tentolouris-Piperas V, Tzanetakos D, Stathopoulos P, Koutsis G, Spyropoulou-Vlachou M, Evangelopoulos ME, Stefanis L, Stavropoulos-Giokas C, Anagnostouli M. HLA-genotyping by Next-Generation-Sequencing reveals shared and unique HLA alleles in two patients with coexisting neuromyelitis optica spectrum disorder and thymectomized myasthenia gravis: immunological implications for mutual aetiopathogenesis? Mult Scler Relat Disord 2022; 63:103858. [DOI: 10.1016/j.msard.2022.103858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/10/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
|
19
|
Frieser D, Pignata A, Khajavi L, Shlesinger D, Gonzalez-Fierro C, Nguyen XH, Yermanos A, Merkler D, Höftberger R, Desestret V, Mair KM, Bauer J, Masson F, Liblau RS. Tissue-resident CD8 + T cells drive compartmentalized and chronic autoimmune damage against CNS neurons. Sci Transl Med 2022; 14:eabl6157. [PMID: 35417189 DOI: 10.1126/scitranslmed.abl6157] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying the chronicity of autoimmune diseases of the central nervous system (CNS) are largely unknown. In particular, it is unclear whether tissue-resident memory T cells (TRM) contribute to lesion pathogenesis during chronic CNS autoimmunity. Here, we observed that a high frequency of brain-infiltrating CD8+ T cells exhibit a TRM-like phenotype in human autoimmune encephalitis. Using mouse models of neuronal autoimmunity and a combination of T single-cell transcriptomics, high-dimensional flow cytometry, and histopathology, we found that pathogenic CD8+ T cells behind the blood-brain barrier adopt a characteristic TRM differentiation program, and we revealed their phenotypic and functional heterogeneity. In the diseased CNS, autoreactive tissue-resident CD8+ T cells sustained focal neuroinflammation and progressive loss of neurons, independently of recirculating CD8+ T cells. Consistently, a large fraction of autoreactive tissue-resident CD8+ T cells exhibited proliferative potential as well as proinflammatory and cytotoxic properties. Persistence of tissue-resident CD8+ T cells in the CNS and their functional output, but not their initial differentiation, were crucially dependent on CD4+ T cells. Collectively, our results point to tissue-resident CD8+ T cells as essential drivers of chronic CNS autoimmunity and suggest that therapies targeting this compartmentalized autoreactive T cell subset might be effective for treating CNS autoimmune diseases.
Collapse
Affiliation(s)
- David Frieser
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Aurora Pignata
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Leila Khajavi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | | | - Carmen Gonzalez-Fierro
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Xuan-Hung Nguyen
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Alexander Yermanos
- Institute of Microbiology, ETH Zurich, 8093 Zurich, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland.,Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Virginie Desestret
- National Reference Center for Paraneoplastic Neurological Syndromes, MeLiS-UCBL-CNRS, INSERM, Hôpital Neurologique, Hospices Civils de Lyon, 69500 Lyon, France
| | - Katharina M Mair
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Frederick Masson
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France
| | - Roland S Liblau
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, CNRS, INSERM, UPS, 31024 Toulouse, France.,Department of Immunology, Toulouse University Hospital, 31300 Toulouse, France
| |
Collapse
|
20
|
Kulkarni B, Cruz-Martins N, Kumar D. Microglia in Alzheimer's Disease: An Unprecedented Opportunity as Prospective Drug Target. Mol Neurobiol 2022; 59:2678-2693. [PMID: 35149973 DOI: 10.1007/s12035-021-02661-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/20/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease (AD) is an ever more common neurodegenerative disease among the elderly, characterized by recurrent neuroinflammation and amyloid beta (Aβ) accumulation in the brain parenchyma. Recent genome-wide association studies (GWAS) have shown a distinct role for the innate immune system in AD, with microglia playing a key role. The function of microglial cells is stringently regulated by the neighboring microenvironment in the brain. Upon interruption in diseases, like AD, it demonstrates neurotoxic and neuroprotective action by M1 (neurotoxic) and M2 (neuroprotective) microglial phenotypes, respectively, in the brain. Microglial cells on activation by complement factors, toll-like receptors, and genetic variants result in Aβ' phagocytosis, synaptic pruning, and reactivation of complement pathway. Recent studies have demonstrated the presence of potential therapeutic targets in microglial cells. Immune receptors revealed on microglia as potential drug targets can be paired immunoglobulin-like type 2 receptor (PILR), CD3358, and triggering receptor expressed on myeloid cells 2 (TREM2), as they can have impact on late-onset AD occurrence and progression. Thus, targeting these receptors can accentuate the beneficial effects of microglial cells required to decelerate the progression of AD. This review emphasizes the microglial phenotypes, its function in AD brain, and potential immunological and therapeutic targets to fight this highly progressive neurodegenerative disorder.
Collapse
Affiliation(s)
- Bhargavi Kulkarni
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed To Be University) Erandawane, Pune, 411038, Maharashtra, India
| | - Natália Cruz-Martins
- Institute of Research and Advanced, Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116, Gandra, PRD, Portugal. .,Faculty of Medicine, University of Porto, Alameda Prof. Hernani Monteiro, 4200-319, Porto, Portugal. .,Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135, Porto, Portugal.
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed To Be University) Erandawane, Pune, 411038, Maharashtra, India.
| |
Collapse
|
21
|
High cell surface expression and peptide binding affinity of HLA-DQA1*05:03, a susceptible allele of neuromyelitis optica spectrum disorders (NMOSD). Sci Rep 2022; 12:106. [PMID: 34997058 PMCID: PMC8742014 DOI: 10.1038/s41598-021-04074-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/13/2021] [Indexed: 11/08/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing autoimmune disease characterized by the presence of pathogenic autoantibodies, anti-aquaporin 4 (AQP4) antibodies. Recently, HLA-DQA1*05:03 was shown to be significantly associated with NMOSD in a Japanese patient cohort. However, the specific mechanism by which HLA-DQA1*05:03 is associated with the development of NMOSD has yet to be elucidated. In the current study, we revealed that HLA-DQA1*05:03 exhibited significantly higher cell surface expression levels compared to other various DQA1 alleles, and that its expression strongly depended on the amino acid sequence of the α1 domain, with a preference for leucine at position 75. Moreover, in silico analysis indicated that the HLA-DQ encoded by HLA-DQA1*05:03 preferentially presents immunodominant AQP4 peptides, and that the peptide major histocompatibility complexes (pMHCs) are more energetically stable in the presence of HLA-DQA1*05:03 than other HLA-DQA1 alleles. In silico 3D structural models were also applied to investigate the validity of the energetic stability of pMHCs. Taken together, our findings indicate that HLA-DQA1*05:03 possesses a distinct property to play a pathogenic role in the development of NMOSD.
Collapse
|
22
|
Abstract
The realization that autoantibodies can contribute to dysfunction of the brain has brought about a paradigm shift in neurological diseases over the past decade, offering up important novel diagnostic and therapeutic opportunities. Detection of specific autoantibodies to neuronal or glial targets has resulted in a better understanding of central nervous system autoimmunity and in the reclassification of some diseases previously thought to result from infectious, 'idiopathic' or psychogenic causes. The most prominent examples, such as aquaporin 4 autoantibodies in neuromyelitis optica or NMDAR autoantibodies in encephalitis, have stimulated an entire field of clinical and experimental studies on disease mechanisms and immunological abnormalities. Also, these findings inspired the search for additional autoantibodies, which has been very successful to date and has not yet reached its peak. This Review summarizes this rapid development at a point in time where preclinical studies have started delivering fundamental new data for mechanistic understanding, where new technologies are being introduced into this field, and - most importantly - where the first specifically tailored immunotherapeutic approaches are emerging.
Collapse
Affiliation(s)
- Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
23
|
Pittock SJ, Zekeridou A, Weinshenker BG. Hope for patients with neuromyelitis optica spectrum disorders - from mechanisms to trials. Nat Rev Neurol 2021; 17:759-773. [PMID: 34711906 DOI: 10.1038/s41582-021-00568-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 02/07/2023]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a rare inflammatory CNS disease that primarily manifests as relapsing episodes of severe optic neuritis and myelitis. Diagnosis of NMOSD is supported by the detection of IgG autoantibodies that target the aquaporin 4 (AQP4) water channel, which, in the CNS, is an astrocyte-specific protein. AQP4 antibody binding leads to AQP4 internalization, complement-dependent and antibody-dependent cellular cytotoxicity, and water channel dysfunction. Cumulative attack-related injury causes disability in NMOSD, so the prevention of attacks is expected to prevent disability accrual. Until recently, no regulator-approved therapies were available for NMOSD. Traditional immunosuppressant therapies, including mycophenolate mofetil, azathioprine and rituximab, were widely used but their benefits have not been assessed in controlled studies. In 2019 and 2020, five phase II and III randomized placebo-controlled trials of four mechanism-based therapies for NMOSD were published and demonstrated that all four effectively prolonged the time to first relapse. All four drugs were monoclonal antibodies: the complement C5 antibody eculizumab, the IL-6 receptor antibody satralizumab, the B cell-depleting antibody inebilizumab, which targets CD19, and rituximab, which targets CD20. We review the pathophysiology of NMOSD, the rationale for the development of these mechanism-based drugs, the methodology and outcomes of the five trials, and the implications of these findings for the treatment of NMOSD.
Collapse
Affiliation(s)
- Sean J Pittock
- Department of Neurology, Mayo Clinic, Rochester, MN, USA. .,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA. .,Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| | - Anastasia Zekeridou
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Brian G Weinshenker
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.,Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
24
|
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
|
25
|
Liu X, Yu Z, Wu Y, Shi S, Yao J, Feng X, Wen D, Shi Z, Zhao Z, Li Y, Zhou H, You C, Lin Y, Yang M. The immune regulatory effects of tetrahedral framework nucleic acid on human T cells via the mitogen-activated protein kinase pathway. Cell Prolif 2021; 54:e13084. [PMID: 34170049 PMCID: PMC8349649 DOI: 10.1111/cpr.13084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Autoimmune diseases are a heterogeneous group of diseases which lose the immunological tolerance to self-antigens. It is well recognized that irregularly provoked T cells participate in the pathological immune responses. As a novel nanomaterial with promising applications, tetrahedral framework nucleic acid (TFNA) nanostructure was found to have immune regulatory effects on T cells in this study. MATERIALS AND METHODS To verify the successful fabrication of TFNA, the morphology of TFNA was observed by atomic force microscopy (AFM) and dynamic light scattering. The regulatory effect of TFNA was evaluated by flow cytometry after cocultured with CD3+ T cells isolated from healthy donors. Moreover, the associated signaling pathways were investigated. Finally, we verified our results on the T cells from patients with neuromyelitis optica spectrum disorder (NMOSD), which is a typical autoimmune disease induced by T cells. RESULTS We revealed the alternative regulatory functions of TFNA in human primary T cells with steady status via the JNK signaling pathway. Moreover, by inhibiting both JNK and ERK phosphorylation, TFNA exhibited significant suppressive effects on IFNγ secretion from provoking T cells without affecting TNF secretion. Similar immune regulatory effects of TFNA were also observed in autoreactive T cells from patients with NMOSD. CONCLUSIONS Overall, our results revealed a potential application of TFNA in regulating the adaptive immune system, as well as shed a light on the treatment of T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Xuyang Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyuan Yu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ya Wu
- Sichuan Cancer Hospital & Institute, Centre for Translational Research in Cancer, Sichuan Cancer Center, Chengdu, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Yao
- Sichuan Cancer Hospital & Institute, Centre for Translational Research in Cancer, Sichuan Cancer Center, Chengdu, China
| | - Xiaorong Feng
- Sichuan Cancer Hospital & Institute, Centre for Translational Research in Cancer, Sichuan Cancer Center, Chengdu, China
| | - Dingke Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ziyan Shi
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengyang Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hongyu Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Mu Yang
- Sichuan Cancer Hospital & Institute, Centre for Translational Research in Cancer, Sichuan Cancer Center, Chengdu, China.,School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
26
|
Schirmer L, Schafer DP, Bartels T, Rowitch DH, Calabresi PA. Diversity and Function of Glial Cell Types in Multiple Sclerosis. Trends Immunol 2021; 42:228-247. [PMID: 33593693 PMCID: PMC7914214 DOI: 10.1016/j.it.2021.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022]
Abstract
Glial subtype diversity is an emerging topic in neurobiology and immune-mediated neurological diseases such as multiple sclerosis (MS). We discuss recent conceptual and technological advances that allow a better understanding of the transcriptomic and functional heterogeneity of oligodendrocytes (OLs), astrocytes, and microglial cells under inflammatory-demyelinating conditions. Recent single cell transcriptomic studies suggest the occurrence of novel homeostatic and reactive glial subtypes and provide insight into the molecular events during disease progression. Multiplexed RNA in situ hybridization has enabled 'mapping back' dysregulated gene expression to glial subtypes within the MS lesion microenvironment. These findings suggest novel homeostatic and reactive glial-cell-type functions both in immune-related processes and neuroprotection relevant to understanding the pathology of MS.
Collapse
Affiliation(s)
- Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| | - Dorothy P Schafer
- Department of Neurobiology and the Brudnik Neuropsychiatric Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Theresa Bartels
- Department of Paediatrics, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - David H Rowitch
- Department of Paediatrics, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Departments of Pediatrics and Neurosurgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Muraki Y, Nishimoto Y, Yamasaki M, Miyakawa S, Sato S. The evaluation of lymph node cell proliferation response by liposomes loaded with major histocompatibility complex class II binding aquaporin 4 antigen peptide. Biosci Biotechnol Biochem 2021; 85:537-544. [PMID: 33624776 DOI: 10.1093/bbb/zbaa084] [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] [Received: 08/18/2020] [Accepted: 11/09/2020] [Indexed: 11/14/2022]
Abstract
Autoimmune responses to aquaporin 4 (AQP4) cause neuromyelitis optica (NMO); thus, specific immunotolerance to this self-antigen could represent a new NMO treatment. We generated the liposome-encapsulated AQP4 peptide 201-220 (p201-220) to induce immunotolerance. Liposomes were generated using phosphatidylserine and the polyglycidol species PG8MG. The in vivo tissue distribution of the liposomes was tested using an ex vivo imaging system. To confirm the antigen presentation capacity of PG8MG liposomes, dendritic cells were treated with PG8MG liposome-encapsulated AQP4 p201-220 (AQP4-PG8MG liposomes). Immunotolerance induction by AQP4-PG8MG liposomes was evaluated using the ex vivo cell proliferation of lymph node cells isolated from AQP4 p201-220-immunized AQP4-deficient mice. Fluorescent dye-labeled PG8MG liposomes were distributed to the lymph nodes. AQP4 p201-220 was presented on dendritic cells. AQP4-PG8MG liposomes were tended to suppress immune responses to AQP4 p201-220. Thus, the encapsulation of AQP4 peptides in PG8MG liposomes represents a new strategy for suppressing autoimmune responses to AQP4.
Collapse
Affiliation(s)
- Yo Muraki
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yutaka Nishimoto
- Pharmaceutical Sciences, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Midori Yamasaki
- T-CiRA, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Shuuichi Miyakawa
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Shuji Sato
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
28
|
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
|
29
|
Asavapanumas N, Tradtrantip L, Verkman AS. Targeting the complement system in neuromyelitis optica spectrum disorder. Expert Opin Biol Ther 2021; 21:1073-1086. [PMID: 33513036 DOI: 10.1080/14712598.2021.1884223] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorder (NMOSD) is characterized by central nervous system inflammation and demyelination. In AQP4-IgG seropositive NMOSD, circulating immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4) cause tissue injury. Compelling evidence supports a pathogenic role for complement activation following AQP4-IgG binding to AQP4. Clinical studies supported the approval of eculizumab, an inhibitor of C5 cleavage, in AQP4-IgG seropositive NMOSD. AREAS COVERED This review covers in vitro, animal models, and human evidence for complement-dependent and complement-independent tissue injury in AQP4-IgG seropositive NMOSD. Complement targets are discussed, including complement proteins, regulators and anaphylatoxin receptors, and corresponding drug candidates. EXPERT OPINION Though preclinical data support a central pathogenic role of complement activation in AQP4-IgG seropositive NMOSD, they do not resolve the relative contributions of complement-dependent vs. complement-independent disease mechanisms such as antibody-dependent cellular cytotoxicity, T cell effector mechanisms, and direct AQP4-IgG-induced cellular injury. The best evidence that complement-dependent mechanisms predominate in AQP4-IgG seropositive NMOSD comes from eculizumab clinical data. Various drug candidates targeting distinct complement effector mechanisms may offer improved safety and efficacy. However, notwithstanding the demonstrated efficacy of complement inhibition in AQP4-IgG seropositive NMOSD, the ultimate niche for complement inhibition is not clear given multiple drug options with alternative mechanisms of action.Abbreviations: AAV2, Adeno-associated virus 2; ADCC, antibody-dependent cellular cytotoxicity; ANCA, antineutrophilic cytoplasmic autoantibody; AQP4, aquaporin-4; AQP4-IgG, AQP4-immunoglobulin G; C1-INH, C1-esterase inhibitor; C3aR, C3a receptor; C4BP, C4 binding protein; C5aR, C5a receptor; CDC, complement-dependent cytotoxicity; CFHR1, complement factor H related 1; CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; EndoS, endoglycosidase S; FHL-1, factor-H-like protein 1; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor protein-1; IgG, immunoglobulin G; IVIG, intravenous human immunoglobulin G; MAC, membrane attack complex; MBL, maltose-binding lectin; MBP, myelin basic protein; MOG, myelin oligodendrocyte glycoprotein; NK cell, natural killer cell; NMOSD, neuromyelitis optica spectrum disorder; OAP, orthogonal arrays of particles; PNH, paroxysmal nocturnal hemoglobinuria.
Collapse
Affiliation(s)
- Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
30
|
Chen T, Lennon VA, Liu YU, Bosco DB, Li Y, Yi MH, Zhu J, Wei S, Wu LJ. Astrocyte-microglia interaction drives evolving neuromyelitis optica lesion. J Clin Invest 2021; 130:4025-4038. [PMID: 32568214 DOI: 10.1172/jci134816] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
Neuromyelitis optica (NMO) is a severe inflammatory autoimmune CNS disorder triggered by binding of an IgG autoantibody to the aquaporin 4 (AQP4) water channel on astrocytes. Activation of cytolytic complement has been implicated as the major effector of tissue destruction that secondarily involves myelin. We investigated early precytolytic events in the evolving pathophysiology of NMO in mice by continuously infusing IgG (NMO patient serum-derived or AQP4-specific mouse monoclonal), without exogenous complement, into the spinal subarachnoid space. Motor impairment and sublytic NMO-compatible immunopathology were IgG dose dependent, AQP4 dependent, and, unexpectedly, microglia dependent. In vivo spinal cord imaging revealed a striking physical interaction between microglia and astrocytes that required signaling from astrocytes by the C3a fragment of their upregulated complement C3 protein. Astrocytes remained viable but lost AQP4. Previously unappreciated crosstalk between astrocytes and microglia involving early-activated CNS-intrinsic complement components and microglial C3a receptor signaling appears to be a critical driver of the precytolytic phase in the evolving NMO lesion, including initial motor impairment. Our results indicate that microglia merit consideration as a potential target for NMO therapeutic intervention.
Collapse
Affiliation(s)
| | - Vanda A Lennon
- Department of Neurology.,Department of Immunology, and.,Department of Laboratory Medicine/Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | | | | - Shihui Wei
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Long-Jun Wu
- Department of Neurology.,Department of Immunology, and.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
31
|
Dao LM, Machule ML, Bacher P, Hoffmann J, Ly LT, Wegner F, Scheffold A, Prüss H. Decreased inflammatory cytokine production of antigen-specific CD4 + T cells in NMDA receptor encephalitis. J Neurol 2021; 268:2123-2131. [PMID: 33442772 PMCID: PMC8179900 DOI: 10.1007/s00415-020-10371-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022]
Abstract
Anti-N-methyl-D-aspartate-receptor (NMDAR) encephalitis is the most common autoimmune encephalitis with psychosis, amnesia, seizures and dyskinesias. The disease is mediated by pathogenic autoantibodies against the NR1 subunit that disrupt NMDAR function. Antibody infusion into mouse brains can recapitulate encephalitis symptoms, while active immunization resulted also in strong T cell infiltration into the hippocampus. However, whether T cells react against NMDAR and their specific contribution to disease development are poorly understood. Here we characterized the ex vivo frequency and phenotype of circulating CD4+ T helper (TH) cells reactive to NR1 protein using antigen-reactive T cell enrichment (ARTE) in 24 patients with NMDAR encephalitis, 13 patients with LGI1 encephalitis and 51 matched controls. Unexpectedly, patients with NMDAR encephalitis had lower frequencies of CD154-expressing NR1-reactive TH cells than healthy controls and produced significantly less inflammatory cytokines. No difference was seen in T cells reactive to the synaptic target LGI1 (Leucine-rich glioma-inactivated 1), ubiquitous Candida antigens or neoantigens, suggesting that the findings are disease-specific and not related to therapeutic immunosuppression. Also, patients with LGI1 encephalitis showed unaltered numbers of LGI1 antigen-reactive T cells. The data reveal disease-specific functional alterations of circulating NMDAR-reactive TH cells in patients with NMDAR encephalitis and challenge the idea that increased pro-inflammatory NMDAR-reactive T cells contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Le-Minh Dao
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Marie-Luise Machule
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Julius Hoffmann
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Lam-Thanh Ly
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.,Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany. .,Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
32
|
Abstract
Optic neuritis (ON) is an inflammatory attack of the optic nerve that leads to visual disability. It is the most common optic neuropathy affecting healthy young adults, most commonly women aged 20-45 years. It can be idiopathic and monophasic or as part of a neurologic disease such as multiple sclerosis with recurrence and cumulative damage. Currently, there is no therapy to repair the damage from optic neuritis. Animal models are an essential tool for the understanding of the pathogenesis of optic neuritis and for the development of potential treatment strategies. Experimental autoimmune encephalomyelitis (EAE) is the most commonly used experimental rodent model for human autoimmune inflammatory demyelinating diseases of the central nervous system (CNS). In this review, we discuss the latest rodent models regarding optic neuritis, focusing on EAE model, and on its recent achievements and developments.
Collapse
Affiliation(s)
- Yael Redler
- Department of Neuro-Ophthalmology, Massachusetts Eye & Ear Infirmary/Harvard Medical School, Boston, MA, United States
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
The contribution of thymic tolerance to central nervous system autoimmunity. Semin Immunopathol 2020; 43:135-157. [PMID: 33108502 PMCID: PMC7925481 DOI: 10.1007/s00281-020-00822-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases of the central nervous system (CNS) are associated with high levels of morbidity and economic cost. Research efforts have previously focused on the contribution of the peripheral adaptive and innate immune systems to CNS autoimmunity. However, a failure of thymic negative selection is a necessary step in CNS-reactive T cells escaping into the periphery. Even with defective thymic or peripheral tolerance, the development of CNS inflammation is rare. The reasons underlying this are currently poorly understood. In this review, we examine evidence implicating thymic selection in the pathogenesis of CNS autoimmunity. Animal models suggest that thymic negative selection is an important factor in determining susceptibility to and severity of CNS inflammation. There are indirect clinical data that suggest thymic function is also important in human CNS autoimmune diseases. Specifically, the association between thymoma and paraneoplastic encephalitis and changes in T cell receptor excision circles in multiple sclerosis implicate thymic tolerance in these diseases. We identify potential associations between CNS autoimmunity susceptibility factors and thymic tolerance. The therapeutic manipulation of thymopoiesis has the potential to open up new treatment modalities, but a better understanding of thymic tolerance in CNS autoimmunity is required before this can be realised.
Collapse
|
34
|
Jarius S, Paul F, Weinshenker BG, Levy M, Kim HJ, Wildemann B. Neuromyelitis optica. Nat Rev Dis Primers 2020; 6:85. [PMID: 33093467 DOI: 10.1038/s41572-020-0214-9] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
Neuromyelitis optica (NMO; also known as Devic syndrome) is a clinical syndrome characterized by attacks of acute optic neuritis and transverse myelitis. In most patients, NMO is caused by pathogenetic serum IgG autoantibodies to aquaporin 4 (AQP4), the most abundant water-channel protein in the central nervous system. In a subset of patients negative for AQP4-IgG, pathogenetic serum IgG antibodies to myelin oligodendrocyte glycoprotein, an antigen in the outer myelin sheath of central nervous system neurons, are present. Other causes of NMO (such as paraneoplastic disorders and neurosarcoidosis) are rare. NMO was previously associated with a poor prognosis; however, treatment with steroids and plasma exchange for acute attacks and with immunosuppressants (in particular, B cell-depleting agents) for attack prevention has greatly improved the long-term outcomes. Recently, a number of randomized controlled trials have been completed and the first drugs, all therapeutic monoclonal antibodies, have been approved for the treatment of AQP4-IgG-positive NMO and its formes frustes.
Collapse
Affiliation(s)
- Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | - Friedemann Paul
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Ho Jin Kim
- Department of Neurology, Research Institute and Hospital of National Cancer Center, Goyang, Korea
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
35
|
Thoman ME, McKarns SC. Metabolomic Profiling in Neuromyelitis Optica Spectrum Disorder Biomarker Discovery. Metabolites 2020; 10:metabo10090374. [PMID: 32961928 PMCID: PMC7570337 DOI: 10.3390/metabo10090374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
There is no specific test for diagnosing neuromyelitis optica spectrum disorder (NMOSD), a disabling autoimmune disease of the central nervous system. Instead, diagnosis relies on ruling out other related disorders with overlapping clinical symptoms. An urgency for NMOSD biomarker discovery is underscored by adverse responses to treatment following misdiagnosis and poor prognosis following the delayed onset of treatment. Pathogenic autoantibiotics that target the water channel aquaporin-4 (AQP4) and myelin oligodendrocyte glycoprotein (MOG) contribute to NMOSD pathology. The importance of early diagnosis between AQP4-Ab+ NMOSD, MOG-Ab+ NMOSD, AQP4-Ab− MOG-Ab− NMOSD, and related disorders cannot be overemphasized. Here, we provide a comprehensive data collection and analysis of the currently known metabolomic perturbations and related proteomic outcomes of NMOSD. We highlight short chain fatty acids, lipoproteins, amino acids, and lactate as candidate diagnostic biomarkers. Although the application of metabolomic profiling to individual NMOSD patient care shows promise, more research is needed.
Collapse
Affiliation(s)
- Maxton E. Thoman
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Susan C. McKarns
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Laboratory of TGF-β Biology, Epigenetics, and Cytokine Regulation, Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Department of Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Correspondence:
| |
Collapse
|
36
|
Hofer LS, Ramberger M, Gredler V, Pescoller AS, Rostásy K, Sospedra M, Hegen H, Berger T, Lutterotti A, Reindl M. Comparative Analysis of T-Cell Responses to Aquaporin-4 and Myelin Oligodendrocyte Glycoprotein in Inflammatory Demyelinating Central Nervous System Diseases. Front Immunol 2020; 11:1188. [PMID: 32625206 PMCID: PMC7311656 DOI: 10.3389/fimmu.2020.01188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022] Open
Abstract
Autoantibodies against aquaporin-4 (AQP4-Ab) and myelin oligodendrocyte glycoprotein (MOG-Ab) are associated with rare central nervous system inflammatory demyelinating diseases like neuromyelitis optica spectrum disorders (NMOSD). Previous studies have shown that not only antibodies, but also autoreactive T-cell responses against AQP4 are present in NMOSD. However, no study has yet analyzed the presence of MOG reactive T-cells in patients with MOG antibodies. Therefore, we compared AQP4 and MOG specific peripheral T-cell response in individuals with AQP4-Ab (n = 8), MOG-Ab (n = 10), multiple sclerosis (MS, n = 8), and healthy controls (HC, n = 14). Peripheral blood mononuclear cell cultures were stimulated with eight AQP4 and nine MOG peptides selected from previous studies and a tetanus toxoid peptide mix as a positive control. Antigen-specific T-cell responses were assessed using the carboxyfluorescein diacetate succinimidyl ester proliferation assay and the detection of granulocyte macrophage colony-stimulating factor (GM-CSF), interferon (IFN)-ɤ and interleukin (IL)-4, IL-6, and IL-17A in cell culture supernatants. Additionally, human leukocyte antigen (HLA)-DQ and HLA-DR genotyping of all participants was performed. We classified a T-cell response as positive if proliferation (measured by a cell division index ≥3) was confirmed by the secretion of at least one cytokine. Reactivity against AQP4 peptides was observed in many groups, but the T-cell response against AQP4 p156-170 was present only in patients with AQP4-Ab (4/8, 50%) and absent in patients with MOG-Ab, MS and HC (corrected p = 0.02). This AQP4 p156-170 peptide specific T-cell response was significantly increased in participants with AQP4-Ab compared to those without [Odds ratio (OR) = 59.00, 95% confidence interval-CI 2.70–1,290.86]. Moreover, T-cell responses against at least one AQP4 peptide were also more frequent in participants with AQP4-Ab (OR = 11.45, 95% CI 1.24–106.05). We did not observe any significant differences for the other AQP4 peptides or any MOG peptide. AQP4-Ab were associated with HLA DQB1*02 (OR = 5.71, 95% CI 1.09–30.07), DRB1*01 (OR = 9.33, 95% CI 1.50–58.02) and DRB1*03 (OR = 6.75, 95% CI = 1.19–38.41). Furthermore, HLA DRB1*01 was also associated with the presence of AQP4 p156-170 reactive T-cells (OR = 31.67, 95% CI 1.30–772.98). To summarize, our findings suggest a role of AQP4-specific, but not MOG-specific T-cells, in NMOSD.
Collapse
Affiliation(s)
- Livia Sophie Hofer
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Melanie Ramberger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria.,Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Viktoria Gredler
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Sophie Pescoller
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kevin Rostásy
- Paediatric Neurology, Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany
| | - Mireia Sospedra
- Department of Neuroimmunology, University of Zurich, Zurich, Switzerland
| | - Harald Hegen
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Andreas Lutterotti
- Department of Neuroimmunology, University of Zurich, Zurich, Switzerland
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
37
|
Agasing AM, Wu Q, Khatri B, Borisow N, Ruprecht K, Brandt AU, Gawde S, Kumar G, Quinn JL, Ko RM, Mao-Draayer Y, Lessard CJ, Paul F, Axtell RC. Transcriptomics and proteomics reveal a cooperation between interferon and T-helper 17 cells in neuromyelitis optica. Nat Commun 2020; 11:2856. [PMID: 32503977 PMCID: PMC7275086 DOI: 10.1038/s41467-020-16625-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Type I interferon (IFN-I) and T helper 17 (TH17) drive pathology in neuromyelitis optica spectrum disorder (NMOSD) and in TH17-induced experimental autoimmune encephalomyelitis (TH17-EAE). This is paradoxical because the prevalent theory is that IFN-I inhibits TH17 function. Here we report that a cascade involving IFN-I, IL-6 and B cells promotes TH17-mediated neuro-autoimmunity. In NMOSD, elevated IFN-I signatures, IL-6 and IL-17 are associated with severe disability. Furthermore, IL-6 and IL-17 levels are lower in patients on anti-CD20 therapy. In mice, IFN-I elevates IL-6 and exacerbates TH17-EAE. Strikingly, IL-6 blockade attenuates disease only in mice treated with IFN-I. By contrast, B-cell-deficiency attenuates TH17-EAE in the presence or absence of IFN-I treatment. Finally, IFN-I stimulates B cells to produce IL-6 to drive pathogenic TH17 differentiation in vitro. Our data thus provide an explanation for the paradox surrounding IFN-I and TH17 in neuro-autoimmunity, and may have utility in predicting therapeutic response in NMOSD.
Collapse
Affiliation(s)
- Agnieshka M Agasing
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, 940 Stanton L. Young Blvd., BMSB 1053, Oklahoma City, OK, 73104, USA
| | - Qi Wu
- Department of Neurology, University of Michigan Medical School, 109 Zina Pitcher Place, Biomedical Research Building Room 4258, Ann Arbor, MI, 48109, USA
| | - Bhuwan Khatri
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Nadja Borisow
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology with Experimental Neurology, Charité Universitätsmedizin, Charitéplatz 1, Berlin, 10117, Germany
| | - Alexander Ulrich Brandt
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Neurology, University of California, Irvine Hall, R105, 252 Health Sciences Rd: 4290, 92697, Irvine, California, USA
| | - Saurabh Gawde
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, 940 Stanton L. Young Blvd., BMSB 1053, Oklahoma City, OK, 73104, USA
| | - Gaurav Kumar
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - James L Quinn
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, 940 Stanton L. Young Blvd., BMSB 1053, Oklahoma City, OK, 73104, USA
| | - Rose M Ko
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, 109 Zina Pitcher Place, Biomedical Research Building Room 4258, Ann Arbor, MI, 48109, USA
| | - Christopher J Lessard
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Friedemann Paul
- NeuroCure Clinical Research Center and Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité Universitätsmedizin, Charitéplatz 1, Berlin, 10117, Germany
| | - Robert C Axtell
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA.
- Department of Microbiology and Immunology, Oklahoma University Health Science Center, 940 Stanton L. Young Blvd., BMSB 1053, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
38
|
Shi Z, Chen H, Du Q, Zhang Y, Zhang Q, Qiu Y, Zhao Z, Wang J, Yang M, Zhou H. IRAK1 polymorphisms are associated with susceptibility to neuromyelitis optica spectrum disorder. Mult Scler Relat Disord 2020; 37:101438. [PMID: 32173002 DOI: 10.1016/j.msard.2019.101438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND X chromosome-linked interleukin-1 receptor-associated kinase (IRAK1) polymorphisms have been demonstrated to be associated with the risks of several autoimmune diseases, such as systemic lupus erythematosus, systemic sclerosis, rheumatoid arthritis, and autoimmune thyroid diseases. However, no studies have investigated the association of IRAK1 polymorphisms with neuromyelitis optica spectrum disorder (NMOSD). This case-control study was performed to determine the correlation between IRAK1 polymorphisms and the risk of NMOSD. METHODS Two single nucleotide polymorphisms (SNPs) rs1059703G>A and rs3027898C>A of IRAK1 were selected and genotyped using SNPscan in a Chinese cohort, including 332 patients with NMOSD and 520 healthy controls. Chi-square tests and logistic regression analyses were used to determine the associations between IRAK1 polymorphisms and the risk of NMOSD. RESULTS Patients with NMOSD showed a lower frequency of the minor allele A of rs1059703 than did controls (Odds ratio [OR] = 0.68; 95% confidence intervals [CI], 0.52-0.88; Pcorr = 0.007). Compared with wild genotype GG of rs1059703, homozygous mutation AA and heterozygous mutation GA were significantly associated with the decreased risk of NMOSD after adjusting for sex and age (adjusted OR = 0.64; 95%CI, 0.49-0.84; Pcorr = 0.002). Similar associations were also observed for IRAK1 rs3027898C>A. Stratification analysis according to sex revealed that the significantly different allele distributions of the two SNPs were mainly found in females. However, IRAK1 polymorphisms were not correlated with aquaporin-4-IgG, onset symptoms, or age at onset. CONCLUSIONS This study is first to demonstrate that X-chromosome-linked IRAK1 polymorphisms are associated with the risk of NMOSD and provide novel insights into the underlying mechanisms of this disease. Further studies are needed to elucidate the function of IRAK1 variants in the pathogenesis of NMOSD and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ziyan Shi
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongxi Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Du
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuhan Qiu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhengyang Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiancheng Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mu Yang
- Department of Basic Research, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hongyu Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
39
|
Cotzomi E, Stathopoulos P, Lee CS, Ritchie AM, Soltys JN, Delmotte FR, Oe T, Sng J, Jiang R, Ma AK, Vander Heiden JA, Kleinstein SH, Levy M, Bennett JL, Meffre E, O'Connor KC. Early B cell tolerance defects in neuromyelitis optica favour anti-AQP4 autoantibody production. Brain 2020; 142:1598-1615. [PMID: 31056665 DOI: 10.1093/brain/awz106] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 11/12/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) constitute rare autoimmune disorders of the CNS that are primarily characterized by severe inflammation of the spinal cord and optic nerve. Approximately 75% of NMOSD patients harbour circulating pathogenic autoantibodies targeting the aquaporin-4 water channel (AQP4). The source of these autoantibodies remains unclear, but parallels between NMOSD and other autoantibody-mediated diseases posit compromised B cell tolerance checkpoints as common underlying and contributing factors. Using a well established assay, we assessed tolerance fidelity by creating recombinant antibodies from B cell populations directly downstream of each checkpoint and testing them for polyreactivity and autoreactivity. We examined a total of 863 recombinant antibodies. Those derived from three anti-AQP4-IgG seropositive NMOSD patients (n = 130) were compared to 733 antibodies from 15 healthy donors. We found significantly higher frequencies of poly- and autoreactive new emigrant/transitional and mature naïve B cells in NMOSD patients compared to healthy donors (P-values < 0.003), thereby identifying defects in both central and peripheral B cell tolerance checkpoints in these patients. We next explored whether pathogenic NMOSD anti-AQP4 autoantibodies can originate from the pool of poly- and autoreactive clones that populate the naïve B cell compartment of NMOSD patients. Six human anti-AQP4 autoantibodies that acquired somatic mutations were reverted back to their unmutated germline precursors, which were tested for both binding to AQP4 and poly- or autoreactivity. While the affinity of mature autoantibodies against AQP4 ranged from modest to strong (Kd 15.2-559 nM), none of the germline revertants displayed any detectable binding to AQP4, revealing that somatic hypermutation is required for the generation of anti-AQP4 autoantibodies. However, two (33.3%) germline autoantibody revertants were polyreactive and four (66.7%) were autoreactive, suggesting that pathogenic anti-AQP4 autoantibodies can originate from the pool of autoreactive naïve B cells, which develops as a consequence of impaired early B cell tolerance checkpoints in NMOSD patients.
Collapse
Affiliation(s)
- Elizabeth Cotzomi
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Panos Stathopoulos
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Casey S Lee
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Alanna M Ritchie
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - John N Soltys
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - Fabien R Delmotte
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Tyler Oe
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Joel Sng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruoyi Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Anthony K Ma
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Steven H Kleinstein
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.,Interdepartmental Program in Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Levy
- Department of Neurology, Johns Hopkins, School of Medicine, Baltimore, MD, USA
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology and Neuroscience Program, University of Colorado, Denver, CO, USA
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin C O'Connor
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
40
|
Liu J, Mori M, Sugimoto K, Uzawa A, Masuda H, Uchida T, Ohtani R, Kuwabara S. Peripheral blood helper T cell profiles and their clinical relevance in MOG-IgG-associated and AQP4-IgG-associated disorders and MS. J Neurol Neurosurg Psychiatry 2020; 91:132-139. [PMID: 31806723 DOI: 10.1136/jnnp-2019-321988] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/21/2019] [Accepted: 11/16/2019] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To investigate the immunological characteristics and their clinical relevance in anti-myelin oligodendrocyte glycoprotein (MOG)-IgG-associated and anti-aquaporin-4 (AQP4)-IgG-associated disorders (MOGAD and AQPAD) and multiple sclerosis (MS). METHODS We measured peripheral blood helper T cell subsets (Th1, Th2, Th17 and regulatory T cell (Treg)) in patients with MOGAD (n=26), AQPAD (n=32) and MS (n=28) in the attack and remission phases by flow cytometry with intracellular cytokine staining. We also studied their correlation with clinical parameters. Ten normal subjects served as healthy controls. RESULTS In all the three disorders, Th17 significantly increased at attack, and downregulated in the remission phases, although still elevated compare with healthy controls. MOGAD and AQPAD patients shared the common T cell profiles, while the extent of Th17 shift was more prominent in AQPAD. Patients with MS showed decreased Th2 than ones with MOGAD and AQPAD at attack. In terms of clinical correlation, MS patients showed that higher Th1 and Th17 proportion was associated with more frequent relapse and more severe clinical disability, whereas in MOGAD, higher Treg was associated with milder clinical severity. In AQPAD, no obvious correlation of Th profiles with clinical manifestation was found. CONCLUSIONS The present study first investigated intracellular cytokine levels among MOGAD, AQPAD and MS. The different patterns and extent of helper T cell profiles could reflect the pathogenesis of each disorders, and may affect disease severity and activity.
Collapse
Affiliation(s)
- Jia Liu
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Masahiro Mori
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuo Sugimoto
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Neurology, Beijing University of Chinese Medicine Affiliated Dongzhimen Hospital, Beijing, China
| | - Akiyuki Uzawa
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroki Masuda
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomohiko Uchida
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ryohei Ohtani
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuwabara
- Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
41
|
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
|
42
|
Cruz-Herranz A, Dietrich M, Hilla AM, Yiu HH, Levin MH, Hecker C, Issberner A, Hallenberger A, Cordano C, Lehmann-Horn K, Balk LJ, Aktas O, Ingwersen J, von Gall C, Hartung HP, Zamvil SS, Fischer D, Albrecht P, Green AJ. Monitoring retinal changes with optical coherence tomography predicts neuronal loss in experimental autoimmune encephalomyelitis. J Neuroinflammation 2019; 16:203. [PMID: 31684959 PMCID: PMC6827223 DOI: 10.1186/s12974-019-1583-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background Retinal optical coherence tomography (OCT) is a clinical and research tool in multiple sclerosis, where it has shown significant retinal nerve fiber (RNFL) and ganglion cell (RGC) layer thinning, while postmortem studies have reported RGC loss. Although retinal pathology in experimental autoimmune encephalomyelitis (EAE) has been described, comparative OCT studies among EAE models are scarce. Furthermore, the best practices for the implementation of OCT in the EAE lab, especially with afoveate animals like rodents, remain undefined. We aimed to describe the dynamics of retinal injury in different mouse EAE models and outline the optimal experimental conditions, scan protocols, and analysis methods, comparing these to histology to confirm the pathological underpinnings. Methods Using spectral-domain OCT, we analyzed the test-retest and the inter-rater reliability of volume, peripapillary, and combined horizontal and vertical line scans. We then monitored the thickness of the retinal layers in different EAE models: in wild-type (WT) C57Bl/6J mice immunized with myelin oligodendrocyte glycoprotein peptide (MOG35–55) or with bovine myelin basic protein (MBP), in TCR2D2 mice immunized with MOG35–55, and in SJL/J mice immunized with myelin proteolipid lipoprotein (PLP139–151). Strain-matched control mice were sham-immunized. RGC density was counted on retinal flatmounts at the end of each experiment. Results Volume scans centered on the optic disc showed the best reliability. Retinal changes during EAE were localized in the inner retinal layers (IRLs, the combination of the RNFL and the ganglion cell plus the inner plexiform layers). In WT, MOG35–55 EAE, progressive thinning of IRL started rapidly after EAE onset, with 1/3 of total loss occurring during the initial 2 months. IRL thinning was associated with the degree of RGC loss and the severity of EAE. Sham-immunized SJL/J mice showed progressive IRL atrophy, which was accentuated in PLP-immunized mice. MOG35–55-immunized TCR2D2 mice showed severe EAE and retinal thinning. MBP immunization led to very mild disease without significant retinopathy. Conclusions Retinal neuroaxonal damage develops quickly during EAE. Changes in retinal thickness mirror neuronal loss and clinical severity. Monitoring of the IRL thickness after immunization against MOG35–55 in C57Bl/6J mice seems the most convenient model to study retinal neurodegeneration in EAE.
Collapse
Affiliation(s)
- Andrés Cruz-Herranz
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hao H Yiu
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Marc H Levin
- Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.,Department of Ophthalmology, Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Christian Cordano
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Klaus Lehmann-Horn
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lisanne J Balk
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Scott S Zamvil
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA.,Program in Immunology, University of California, San Francisco, San Francisco, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Ari J Green
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA. .,Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
43
|
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
|
44
|
Intradermal vaccination prevents anti-MOG autoimmune encephalomyelitis in macaques. EBioMedicine 2019; 47:492-505. [PMID: 31492559 PMCID: PMC6796575 DOI: 10.1016/j.ebiom.2019.08.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 12/28/2022] Open
Abstract
Background Autoimmune demyelinating diseases (ADD) are a major cause of neurological disability due to autoreactive cellular and humoral immune responses against brain antigens. A cure for chronic ADD could be obtained by appropriate immunomodulation. Methods We implemented a preclinical scheme to foster immune tolerance to myelin oligodendrocyte glycoprotein (MOG), in a cynomolgus-macaque model of experimental autoimmune encephalomyelitis (EAE), in which administration of recombinant human MOG (rhMOG) elicits brain inflammation mediated by MOG-autoreactive CD4+ lymphocytes and anti-MOG IgG. For immunotherapy, we used a recombinant antibody (Ab) directed against the dendritic cell-asialoglycoprotein receptor (DC-ASGPR) fused either to MOG or a control antigen PSA (prostate-specific antigen). Findings rhMOG and the anti-DC-ASGPR-MOG were respectively detected in CD1a+ DCs or CD163+ cells in the skin of macaques. Intradermal administration of anti-DC-ASGPR-MOG, but not control anti-DC-ASGPR-PSA, was protective against EAE. The treatment prevented the CD4+ T cell activation and proinflammatory cytokine production observed in controls. Moreover, the administration of anti-DC-ASGPR-MOG induced MOG-specific CD4+CD25+FOXP3+CD39+ regulatory lymphocytes and favoured an upsurge in systemic TGFβ and IL-8 upon rhMOG re-administration in vivo. Interpretation We show that the delivery of an anti-DC-ASGPR-MOG allows antigen-specific adaptive immune modulation to prevent the breach of immune tolerance to MOG. Our findings pave the way for therapeutic vaccines for long-lasting remission to grave encephalomyelitis with identified autoantigens, such as ADD associated with anti-MOG autoantibodies. Fund Work supported by the French ANR (ANR-11-INBS-0008 and ANR-10-EQPX-02-01), NIH (NIH 1 R01 AI 105066), the Baylor Scott and White Healthcare System funding and Roche Research Collaborative grants.
Collapse
|
45
|
Abstract
The contributions of the peripheral adaptive and innate immune systems to CNS autoimmunity have been extensively studied. However, the role of thymic selection in these conditions is much less well understood. The thymus is the primary lymphoid organ for the generation of T cells; thymic mechanisms ensure that cells with an overt autoreactive specificity are eliminated before they emigrate to the periphery and control the generation of thymic regulatory T cells. Evidence from animal studies demonstrates that thymic T cell selection is important for establishing tolerance to autoantigens. However, there is a considerable knowledge gap regarding the role of thymic selection in autoimmune conditions of the human CNS. In this Review, we critically examine the current body of experimental evidence for the contribution of thymic tolerance to CNS autoimmune diseases. An understanding of why dysfunction of either thymic or peripheral tolerance mechanisms rarely leads to CNS inflammation is currently lacking. We examine the potential of de novo T cell formation and thymic selection as novel therapeutic avenues and highlight areas for future study that are likely to make these targets the focus of future treatments.
Collapse
|
46
|
Chang VTW, Chang HM. Review: Recent advances in the understanding of the pathophysiology of neuromyelitis optica spectrum disorder. Neuropathol Appl Neurobiol 2019; 46:199-218. [PMID: 31353503 DOI: 10.1111/nan.12574] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
Neuromyelitis optica is an autoimmune inflammatory disorder of the central nervous system that preferentially targets the spinal cord and optic nerve. Following the discovery of circulating antibodies against the astrocytic aquaporin 4 (AQP4) water channel protein, recent studies have expanded our knowledge of the unique complexities of the pathogenesis of neuromyelitis optica and its relationship with the immune response. This review describes and summarizes the recent advances in our understanding of the molecular mechanisms underlying neuromyelitis optica disease pathology and examines their potential as therapeutic targets. Additionally, we update the most recent research by proposing major unanswered questions regarding how peripheral AQP4 antibodies are produced and their entry into the central nervous system, the causes of AQP4-IgG-seronegative disease, why peripheral AQP4-expressing organs are spared from damage, and the impact of this disease on pregnancy.
Collapse
Affiliation(s)
- V T W Chang
- St George's, University of London, London, UK
| | - H-M Chang
- Department of Obstetrics and Gynaecology, University of British Columbia and BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
47
|
Hillebrand S, Schanda K, Nigritinou M, Tsymala I, Böhm D, Peschl P, Takai Y, Fujihara K, Nakashima I, Misu T, Reindl M, Lassmann H, Bradl M. Circulating AQP4-specific auto-antibodies alone can induce neuromyelitis optica spectrum disorder in the rat. Acta Neuropathol 2019; 137:467-485. [PMID: 30564980 PMCID: PMC6514074 DOI: 10.1007/s00401-018-1950-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022]
Abstract
It is well established that the binding of pathogenic aquaporin-4 (AQP4)-specific autoantibodies to astrocytes may initiate a cascade of events culminating in the destruction of these cells and in the formation of large tissue-destructive lesions typical for patients with neuromyelitis optica spectrum disorders (NMOSD). To date, not a single experimental study has shown that the systemic presence of the antibody alone can induce any damage to the central nervous system (CNS), while pathological studies on brains of NMOSD patients suggested that there might be ways for antibody entry and subsequent tissue damage. Here, we systemically applied a highly pathogenic, monoclonal antibody with high affinity to AQP4 over prolonged period of time to rats, and show that AQP4-abs can enter the CNS on their own, via circumventricular organs and meningeal or parenchymal blood vessels, that these antibodies initiate the formation of radically different lesions with AQP4 loss, depending on their mode and site of entry, and that lesion formation is much more efficient in the presence of encephalitogenic T-cell responses. We further demonstrate that the established tissue-destructive lesions trigger the formation of additional lesions by short and far reaching effects on blood vessels and their branches, and that AQP4-abs have profound effects on the AQP4 expression in peripheral tissues which counter-act possible titer loss by antibody absorption outside the CNS. Cumulatively, these data indicate that directly induced pathological changes caused by AQP4-abs inside and outside the CNS are efficient drivers of disease evolution in seropositive organisms.
Collapse
Affiliation(s)
- Sophie Hillebrand
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Kathrin Schanda
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Magdalini Nigritinou
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Irina Tsymala
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Denise Böhm
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Patrick Peschl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuo Fujihara
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Nakashima
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
48
|
Brill L, Lavon I, Vaknin-Dembinsky A. Foxp3+ regulatory T cells expression in neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2019; 30:114-118. [PMID: 30771576 DOI: 10.1016/j.msard.2019.01.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Alterations in the frequency or function of regulatory T cells (Tregs), which play a critical role in the maintenance of peripheral immune tolerance, are known to be involved in the pathogenesis of several autoimmune diseases. Neuromyelitis optica spectrum disorders (NMOSD) are autoimmune inflammatory diseases of the central nervous system (CNS), of which the etiology and mechanisms underlying its development are not completely understood. Although there is increasing evidence for the involvement of effector T cells in NMOSD, no data are available regarding the role of Tregs in its pathogenesis. AIM The aim of this study was to investigate the mRNA expression level of regulatory T cell genes in NMOSD. METHODS We used gene expression array and RT-PCR analysis to study Treg cell genes in NMOSD RESULTS: A distinctive Treg gene signature in the peripheral blood of NMOSD patients is described, as well as significantly decreased FoxP3 mRNA expression in the peripheral blood mononuclear cells (PBMCs) of the patients vs that in the healthy controls (HCs) (NMOSD,1.8RQ vs HC, 6.8RQ, p = 0.01). CONCLUSIONS The present study shows downregulation at the mRNA expression level of a Treg key transcription factor FoxP3, in NMOSD. Exploration of Tregs function and interconnections in the peripheral immune system should advance our understanding of NMOSD pathogenesis.
Collapse
Affiliation(s)
- Livnat Brill
- Department of Neurology and the Multiple Sclerosis Center, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University, Ein Karem, P.O.B. 12000, Jerusalem 91120, Israel
| | - Iris Lavon
- Department of Neurology and the Multiple Sclerosis Center, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University, Ein Karem, P.O.B. 12000, Jerusalem 91120, Israel; Leslie and Michael Center for Neuro-oncology, Hadassah- Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and the Multiple Sclerosis Center, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University, Ein Karem, P.O.B. 12000, Jerusalem 91120, Israel.
| |
Collapse
|
49
|
Duan T, Smith AJ, Verkman AS. Complement-dependent bystander injury to neurons in AQP4-IgG seropositive neuromyelitis optica. J Neuroinflammation 2018; 15:294. [PMID: 30348195 PMCID: PMC6198534 DOI: 10.1186/s12974-018-1333-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/15/2018] [Indexed: 01/14/2023] Open
Abstract
Background Aquaporin-4-immunoglobulin G (AQP4-IgG) seropositive neuromyelitis optica spectrum disorder (herein called NMO) is an autoimmune disease of the central nervous system in which AQP4-IgG binding to AQP4 on astrocytes results in complement-dependent astrocyte injury and secondary inflammation, demyelination, and neuron loss. We previously reported evidence for a complement bystander mechanism for early oligodendrocyte injury in NMO. Herein, we tested the hypothesis that complement bystander injury, which involves diffusion to nearby cells of activated soluble complement components from complement-injured astrocytes, is a general phenomenon that may contribute to neuronal injury in NMO. Methods Primary cocultures of rat astrocytes and cortical neurons were established to study complement-dependent cell death after exposure to AQP4-IgG and complement. In animal experiments, AQP4-IgG was delivered to adult rats by intracerebral injection. Cell cultures and rat brain were studied by immunofluorescence. Results In primary astrocyte-neuron cocultures, addition of AQP4-IgG and complement resulted in death of neurons nearby astrocytes. Deposition of complement membrane attack complex C5b-9 was seen on neurons nearby astrocytes, whereas C1q, the initiating protein in the complement pathway, was seen only on astrocytes. Neuron death was not seen with a complement inhibitor, with C1q- or C6-depleted complement, in pure neuron cultures exposed to AQP4-IgG and complement or in cocultures exposed to an astrocyte toxin. Intracerebral injection in rats of AQP4-IgG and a fixable dead cell fluorescent marker produced death of neurons near astrocytes, with C5b-9 deposition. Neuron death was not seen in rats receiving a complement inhibitor or in AQP4-IgG-injected AQP4 knockout rats. Conclusion These results support a novel mechanism for early neuron injury in NMO and provide evidence that complement bystander injury may be a general phenomenon for brain cell injury following AQP4-IgG-targeted astrocyte death. Electronic supplementary material The online version of this article (10.1186/s12974-018-1333-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tianjiao Duan
- Departments of Medicine and Physiology, University of California, 1246 Health Sciences East Tower, 513 Parnassus Ave, San Francisco, CA, 94143-0521, USA.,Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Alex J Smith
- Departments of Medicine and Physiology, University of California, 1246 Health Sciences East Tower, 513 Parnassus Ave, San Francisco, CA, 94143-0521, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, 1246 Health Sciences East Tower, 513 Parnassus Ave, San Francisco, CA, 94143-0521, USA.
| |
Collapse
|
50
|
Cao X, Xu H, Feng W, Su D, Xiao M. Deletion of aquaporin-4 aggravates brain pathology after blocking of the meningeal lymphatic drainage. Brain Res Bull 2018; 143:83-96. [PMID: 30347264 DOI: 10.1016/j.brainresbull.2018.10.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 01/08/2023]
Abstract
The glymphatic pathway and meningeal lymphatic vessels are involved in clearance of metabolic macromolecules from the brain. However, the functional interaction between the two systems in the maintenance of brain homeostasis remains unclear. Here we reported that deletion of aquaporin-4 (AQP4), a functional regulator of glymphatic clearance, aggravated brain pathology of 3 month-old mice after blocking of the meningeal lymphatic drainage for 2 weeks via ligation of the deep cervical lymphatic nodes (LdcLNs). LdcLNs increased total and phosphorylated Tau protein levels in the hippocampus of both genotype mice, but increased hippocampal amyloid beta 1-40 and 1-42 levels only in AQP4 null mice, with up-regulation of beta-site amyloid precursor protein-cleaving enzyme 1 and down-regulation of insulin degrading enzyme. Consistently, LdcLNs caused microglial reactivity and activation of nod-like receptor protein-3 inflammasomes in the AQP4 null hippocampus. These mice also showed hippocampal neuronal apoptosis and declines in exploring and cognitive abilities. Deletion of AQP4, but not LdcLNs, increased brain water content. Together, these findings have revealed respective and interactive roles of the glymphatic system and the dural lymphatic system in maintaining amyloid beta, Tau proteins and water homeostasis in the brain, helping to understand the pathogenesis of neurological diseases associated with mis-accumulation of brain macromolecules.
Collapse
Affiliation(s)
- Xuejin Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China; Key Laboratory for Aging & Disease, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, China
| | - Hanrong Xu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China; Key Laboratory for Aging & Disease, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, China
| | - Dongyuan Su
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China; Key Laboratory for Aging & Disease, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|