1
|
Xu L, Xu H, Tang C. Aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders: progress of experimental models based on disease pathogenesis. Neural Regen Res 2025; 20:354-365. [PMID: 38819039 PMCID: PMC11317952 DOI: 10.4103/nrr.nrr-d-23-01325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/18/2023] [Accepted: 12/19/2023] [Indexed: 06/01/2024] Open
Abstract
Neuromyelitis optica spectrum disorders are neuroinflammatory demyelinating disorders that lead to permanent visual loss and motor dysfunction. To date, no effective treatment exists as the exact causative mechanism remains unknown. Therefore, experimental models of neuromyelitis optica spectrum disorders are essential for exploring its pathogenesis and in screening for therapeutic targets. Since most patients with neuromyelitis optica spectrum disorders are seropositive for IgG autoantibodies against aquaporin-4, which is highly expressed on the membrane of astrocyte endfeet, most current experimental models are based on aquaporin-4-IgG that initially targets astrocytes. These experimental models have successfully simulated many pathological features of neuromyelitis optica spectrum disorders, such as aquaporin-4 loss, astrocytopathy, granulocyte and macrophage infiltration, complement activation, demyelination, and neuronal loss; however, they do not fully capture the pathological process of human neuromyelitis optica spectrum disorders. In this review, we summarize the currently known pathogenic mechanisms and the development of associated experimental models in vitro, ex vivo, and in vivo for neuromyelitis optica spectrum disorders, suggest potential pathogenic mechanisms for further investigation, and provide guidance on experimental model choices. In addition, this review summarizes the latest information on pathologies and therapies for neuromyelitis optica spectrum disorders based on experimental models of aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders, offering further therapeutic targets and a theoretical basis for clinical trials.
Collapse
Affiliation(s)
- Li Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
2
|
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
|
3
|
Wang S, Xue M, Wang J, Wu R, Shao Y, Luo K, Liu J, Zhu M. Effects of intravenous pulse methylprednisolone in neuromyelitis optica during the acute phase. Ann Clin Transl Neurol 2024; 11:2731-2744. [PMID: 39222472 PMCID: PMC11514921 DOI: 10.1002/acn3.52188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/24/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is an anti-aquaporin 4 (anti-AQP4) autoantibodies-mediated idiopathic inflammatory demyelinating disease of the central nervous system. While intravenous pulse methylprednisolone (IVMP) is the recommended initial treatment option for acute onset NMOSD, its therapeutic mechanism remains unclear. We hypothesized that IVMP would reduce the expression of pro-inflammatory factors and increase the resolution of inflammation in patients with NMOSD. METHODS Mendelian randomization (MR) analysis was used to screen meaningful inflammatory and resolution factors for inclusion. Three MR methods with inverse variance weighting (IVW) were primarily used to identify positive results. Interleukin (IL)-10, IL-1β, IL-6, C-X-C motif chemokine ligand 12 (CXCL12), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) were screened from 41 inflammatory factors, and resolvin D1 (RvD1), maresin 1 (MaR1), and lipoxin A4 (LXA4) were screened from 6 resolution markers for inclusion. Subsequently, 12 patients with NMOSD were enrolled and treated with IVMP. Serum levels of the aforementioned inflammatory and resolution markers were measured by enzyme-linked immunosorbent assay before and after IVMP treatment. RESULTS High levels of TRAIL, CXCL12, and IL-1β were associated with an increased risk of NMOSD (TRAIL: odds ratio [OR], 1.582; 95% confidence interval [CI], 1.003-2.495; CXCL12: OR, 3.610; 95% CI, 1.011-12.889; IL-1β: OR, 4.500; 95% CI, 1.129-17.927). High levels of RvD1, MaR1, and LXA4 were associated with a reduced risk of NMOSD (RvD1: OR, 0.725; 95% CI, 0.538-0.976; MaR1: OR, 0.985; 95% CI, 0.970-0.999; LXA4: OR, 0.849; 95% CI, 0.727-0.993). Among patients with NMOSD, serum levels of IL-6, CXCL12, and TRAIL significantly decreased following IVMP treatment, compared with pretreatment levels, while levels of IL-1β, LXA4, and MaR1 significantly increased after IVMP treatment (p < 0.05). A significant positive correlation was observed between CXCL12 levels and Expanded Disability Status Scale (EDSS) scores (r = 0.451, p < 0.05). CONCLUSION Several systemic inflammatory regulators associated with the pathogenesis of NMOSD were identified. The protective roles of LXA4 and MaR1 may be indispensable components of glucocorticoid treatment. Therefore, the use of resolution markers may be a potential strategy for improving central nervous system injury in individuals with NMOSD.
Collapse
Affiliation(s)
- Shengnan Wang
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Mengru Xue
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Jianglong Wang
- First Operating RoomThe First Hospital of Jilin UniversityChangchunChina
| | - Rui Wu
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Yanqing Shao
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Ke Luo
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Jiacheng Liu
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Mingqin Zhu
- Department of Neurology, Neuroscience CenterThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
4
|
Kim M, Kim WS, Cha H, Kim B, Kwon YN, Kim SM. Early involvement of peripherally derived monocytes in inflammation in an NMO-like mouse model. Sci Rep 2024; 14:1177. [PMID: 38216632 PMCID: PMC10786844 DOI: 10.1038/s41598-024-51759-4] [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: 11/01/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune inflammatory disease that primarily affects the optic nerve and spinal cord within the central nervous system (CNS). Acute astrocyte injury caused by autoantibodies against aquaporin 4 (NMO-IgG) is a well-established key factor in the pathogenesis, ultimately leading to neuronal damage and patient disability. In addition to these humoral immune processes, numerous innate immune cells were found in the acute lesions of NMO patients. However, the origin and function of these innate immune cells remain unclear in NMO pathogenesis. Therefore, this study aims to analyze the origin and functions of these innate immune cells in an NMO-like mouse model and evaluate their role in the pathophysiology of NMO. The expression of Tmem119 on Iba1 + cells in brain tissue disappeared immediately after the injection of NMO-IgG + human complement mixture, while the expression of P2ry12 remained well-maintained at 1 day after injection. Based on these observations, it was demonstrated that monocytes infiltrate the brain during the early stages of the pathological process and are closely associated with the inflammatory response through the expression of the proinflammatory cytokine IL-1β. Understanding the variations in the expression patterns of P2ry12, Tmem119, and other markers could be helpful in distinguishing between these cell types and further analyzing their functions. Therefore, this research may contribute to a better understanding of the mechanisms and potential treatments for NMO.
Collapse
Affiliation(s)
- Moonhang Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea.
| | - Won Seok Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Hyeuk Cha
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Boram Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082, Republic of Korea
| | - Young Nam Kwon
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sung Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
5
|
Nakamura N, Ishikawa H, Matsuyama H, Shindo A, Kishida D, Maeda M, Tomimoto H. Neuromyelitis optica spectrum disorder with a familial Mediterranean fever gene E84K mutation. J Neurol 2023; 270:4529-4532. [PMID: 37133537 DOI: 10.1007/s00415-023-11731-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/04/2023]
Affiliation(s)
- Naoko Nakamura
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City, Mie, 514-8507, Japan.
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City, Mie, 514-8507, Japan.
| | - Hirofumi Matsuyama
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City, Mie, 514-8507, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City, Mie, 514-8507, Japan
| | - Dai Kishida
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Nagano, Japan
| | - Masayuki Maeda
- Department of Neuroradiology, Mie University Graduate School of Medicine, Tsu City, Mie, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu City, Mie, 514-8507, Japan
| |
Collapse
|
6
|
Wang Y, Bian J, Yao M, Du L, Xu Y, Chang H, Cong H, Wei Y, Xu W, Wang H, Zhang X, Geng X, Yin L. Targeting chemoattractant chemokine (C-C motif) ligand 2 derived from astrocytes is a promising therapeutic approach in the treatment of neuromyelitis optica spectrum disorders. Front Immunol 2023; 14:1144532. [PMID: 37056770 PMCID: PMC10086366 DOI: 10.3389/fimmu.2023.1144532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction Aquaporin-4 immunoglobulin G (AQP4-IgG)-induced astrocytes injury is a key mechanism in the pathogenesis of neuromyelitis spectrum disorder (NMOSD), and although CCL2 is involved, its specific role has not been reported. We aimed to further investigate the role and potential mechanisms of CCL2 in AQP4-IgG-induced astrocyte injury. Methods First, we evaluated CCL2 levels in paired samples of subject patients by automated microfluidic platform, Ella®. Second, we knock down astrocyte's CCL2 gene in vitro and in vivo to define the function of CCL2 in AQP4-IgG-induced astrocyte injury. Third, astrocyte injury and brain injury in live mice were assessed by immunofluorescence staining and 7.0T MRI, respectively. Western blotting and high-content screening were conducted to clarify the activation of inflammatory signaling pathways, and changes in CCL2 mRNA and cytokine/chemokines were measured by qPCR technique and flow cytometry, respectively. Results There were greatly higher CSF-CCL2 levels in NMOSD patients than that in other non-inflammatory neurological diseases (OND) groups. Blocking astrocyte CCL2 gene expression can efficiently mitigate AQP4-IgG-induced damage in vitro and in vivo. Interestingly, prevention of CCL2 expression could decrease other inflammatory cytokines released, including IL-6 and IL-1β. Our data suggest that CCL2 involves in the initiation and plays a pivotal role in AQP4-IgG-damaged astrocytes. Discussion Our results indicate that CCL2 may serve as a promising candidate target for inflammatory disorder therapy, including NMOSD.
Collapse
Affiliation(s)
- Yupeng Wang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Bian
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mengyuan Yao
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Li Du
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yun Xu
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Haoxiao Chang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hengri Cong
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuzhen Wei
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wangshu Xu
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huabing Wang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xinghu Zhang
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing, China
| | - Linlin Yin
- Department of Neuroinfection and Neuroimmunology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
7
|
The Pathological Activation of Microglia Is Modulated by Sexually Dimorphic Pathways. Int J Mol Sci 2023; 24:ijms24054739. [PMID: 36902168 PMCID: PMC10003784 DOI: 10.3390/ijms24054739] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Microglia are the primary immunocompetent cells of the central nervous system (CNS). Their ability to survey, assess and respond to perturbations in their local environment is critical in their role of maintaining CNS homeostasis in health and disease. Microglia also have the capability of functioning in a heterogeneous manner depending on the nature of their local cues, as they can become activated on a spectrum from pro-inflammatory neurotoxic responses to anti-inflammatory protective responses. This review seeks to define the developmental and environmental cues that support microglial polarization towards these phenotypes, as well as discuss sexually dimorphic factors that can influence this process. Further, we describe a variety of CNS disorders including autoimmune disease, infection, and cancer that demonstrate disparities in disease severity or diagnosis rates between males and females, and posit that microglial sexual dimorphism underlies these differences. Understanding the mechanism behind differential CNS disease outcomes between men and women is crucial in the development of more effective targeted therapies.
Collapse
|
8
|
Schroeder-Castagno M, Del Rio-Serrato A, Wilhelm A, Romero-Suárez S, Schindler P, Alvarez-González C, Duchow AS, Bellmann-Strobl J, Ruprecht K, Hastermann M, Grütz G, Wildemann B, Jarius S, Schmitz-Hübsch T, Paul F, Infante-Duarte C. Impaired response of blood neutrophils to cell-death stimulus differentiates AQP4-IgG-seropositive NMOSD from MOGAD. J Neuroinflammation 2022; 19:239. [PMID: 36183103 PMCID: PMC9526338 DOI: 10.1186/s12974-022-02600-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In neuromyelitis optica spectrum disorders (NMOSD) and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), neutrophils are found in CNS lesions. We previously demonstrated that NMOSD neutrophils show functional deficiencies. Thus, we hypothesized that neutrophil accumulation in the CNS may be facilitated by impairments affecting mechanisms of neutrophil death. OBJECTIVE To evaluate cell death in blood neutrophils from aquaporin-4 (AQP4)-IgG-seropositive NMOSD and MOGAD patients as well as matched healthy controls (HC) using in vitro assays. METHODS Twenty-eight AQP4 + NMOSD and 19 MOGAD patients in stable disease phase as well as 45 age- and sex-matched HC were prospectively recruited. To induce cell death, isolated neutrophils were cultured with/without phorbol 12-myristate 13-acetate (PMA). Spontaneous and PMA-induced NETosis and apoptosis were analyzed using 7-AAD and annexin-V by flow cytometry. Caspase-3 was assessed by western blot. Myeloperoxidase-DNA complexes (MPO-DNA), MPO and elastase were evaluated by ELISA, and cell-free DNA (cfDNA) by a fluorescence-based assay. Reactive oxygen species (ROS) were evaluated by a dihydrorhodamine 123-based cytometric assay. Serum GM-CSF, IL-6, IL-8, IL-15, TNF-ɑ and IL-10 were evaluated by multiplex assays, and neurofilament light chain (NfL) by single-molecule array assay. RESULTS In response to PMA, neutrophils from AQP4 + NMOSD but not from MOGAD patients showed an increased survival, and subsequent reduced cell death (29.6% annexin V+ 7-AAD+) when compared to HC (44.7%, p = 0.0006). However, AQP4 + NMOSD also showed a mild increase in annexin V+ 7-AAD- early apoptotic neutrophils (24.5%) compared to HC (20.8%, p = 0.048). PMA-induced reduction of caspase-3 activation was more pronounced in HC (p = 0.020) than in AQP4 + NMOSD neutrophils (p = 0.052). No differences were observed in neutrophil-derived MPO-DNA or serum levels of MPO, elastase, IL-6, IL-8 and TNF-ɑ. IL-15 levels were increased in both groups of patients. In AQP4 + NMOSD, an increase in cfDNA, GM-CSF and IL-10 was found in serum. A positive correlation among cfDNA and NfL was found in AQP4 + NMOSD. CONCLUSIONS AQP4 + NMOSD neutrophils showed an increased survival capacity in response to PMA when compared to matched HC neutrophils. Although the data indicate that the apoptotic but not the NETotic response is altered in these neutrophils, additional evaluations are required to validate this observation.
Collapse
Affiliation(s)
- Maria Schroeder-Castagno
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Campus Berlin-Buch GmbH, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Alba Del Rio-Serrato
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Campus Berlin-Buch GmbH, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Andreas Wilhelm
- BIH Center for Regenerative Therapies (BCRT) Charité- Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Core Unit Immunocheck-Biomarker Immunologisches Studienlabor (ISL), Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Silvina Romero-Suárez
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, 68167, Mannheim, Germany
| | - Patrick Schindler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Campus Berlin-Buch GmbH, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,Department of Neurology, Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Cesar Alvarez-González
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany.,Neurologic Clinic and Policlinic, Departments of Medicine, University Hospital Basel & RC2NB - Research Center for Clinical Neuroimmunology and Neuroscience, University of Basel, Basel, Switzerland
| | - Ankelien-Solveig Duchow
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Judith Bellmann-Strobl
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Klemens Ruprecht
- Department of Neurology, Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Maria Hastermann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany.,Department of Neurology, Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Gerald Grütz
- BIH Center for Regenerative Therapies (BCRT) Charité- Humboldt-Universität Zu Berlin and Berlin Institute of Health, Institute for Medical Immunology, Core Unit Immunocheck-Biomarker Immunologisches Studienlabor (ISL), Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Sven Jarius
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Tanja Schmitz-Hübsch
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany.,Department of Neurology, Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Friedemann Paul
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, NeuroCure Clinical Research Center, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany.,Department of Neurology, Charité-Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117, Berlin, Germany
| | - Carmen Infante-Duarte
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, ECRC Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité-Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany. .,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Campus Berlin-Buch GmbH, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
9
|
Stathopoulos P, Dalakas MC. The role of complement and complement therapeutics in neuromyelitis optica spectrum disorders. Expert Rev Clin Immunol 2022; 18:933-945. [PMID: 35899480 DOI: 10.1080/1744666x.2022.2105205] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorders (NMOSD) are characterized in the majority of cases by the presence of IgG1 autoantibodies against aquaporin 4 (AQP4) and myelin-oligodendrocyte glycoprotein (MOG), both capable of activating complement. AREAS COVERED We review evidence of complement involvement in NMOSD pathophysiology from pathological, in vitro, in vivo, human studies, and clinical trials. EXPERT OPINION In AQP4 NMOSD, complement deposition is a prominent pathological feature, while in vitro and in vivo studies have demonstrated complement-dependent pathogenicity of AQP4 antibodies. Consistent with these studies, the anti-C5 monoclonal antibody eculizumab was remarkably effective and safe in a phase 2/3 trial of AQP4-NMOSD patents leading to FDA-approved indication. Several other anti-complement agents, either approved or in trials for other neuro-autoimmunities, like myasthenia, CIDP, and GBS, are also relevant to NMOSD generating an exciting group of evolving immunotherapies. Limited but compelling in vivo and in vitro data suggest that anti-complement therapeutics may be also applicable to a subset of MOG NMOSD patients with severe disease. Overall, anticomplement agents, along with the already approved anti-IL6 and anti-CD19 monoclonal antibodies sartralizumab and inebilizumab, are rapidly changing the therapeutic algorithm in NMOSD, a previously difficult-to-treat autoimmune neurological disorder.
Collapse
Affiliation(s)
- Panos Stathopoulos
- Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.,Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Zhang J, Li Y, Zhou Y, Wang K, Pan C, Zhao Y, Xie H, Duan R, Gong Z, Jia Y. Monocyte to High-Density Lipoprotein Ratio: A Novel Predictive Marker of Disease Severity and Prognosis in Patients With Neuromyelitis Optica Spectrum Disorders. Front Neurol 2021; 12:763793. [PMID: 34777231 PMCID: PMC8580507 DOI: 10.3389/fneur.2021.763793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023] Open
Abstract
Background and Purpose: To investigate the association of monocyte to high-density lipoprotein ratio (MHR) with disease severity and prognosis in patients with neuromyelitis optica spectrum disorders (NMOSD). Methods: This retrospective study included 125 patients with NMOSD. Demographic and clinical parameters, including the MHR, were assessed. The initial Expanded Disability Status Scale (EDSS) score and relapse rate were used to evaluate disease severity and prognosis, respectively. Correlations between MHR and disease severity and relapse rate were analyzed. The predictive value of MHR for prognosis was evaluated using receiver operating characteristic (ROC) curve analysis. Results: Compared with the low MHR group, the initial EDSS score (median 4.5 vs. 5.5%, P = 0.025) and relapse rate (51.61 vs. 30.16%, P = 0.015) were significantly higher in the high MHR group. MHR was positively correlated with the initial EDSS score (r = 0.306, P = 0.001). Multivariate analysis showed that MHR was significantly associated with severity (odds ratio = 7.90, 95% confidence interval [CI] = 1.08–57.82, P = 0.041), and it was a significant predictor of disease prognosis (hazard ratio = 3.12, 95% CI = 1.02–9.53, P = 0.046). The median relapse interval of the high MHR group was 24.40 months. When the MHR was higher than 0.565, the risk of relapse was high [sensitivity, 33.3%; specificity, 91.9%; area under the ROC curve, 0.642 (95% CI = 0.54–0.74, P = 0.007)]. Conclusion: MHR is a novel predictive marker of disease severity and prognosis in patients with NMOSD. Early monitoring and reduction of MHR may allow earlier intervention and improved prognosis.
Collapse
Affiliation(s)
- Jinwei Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanfei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongyan Zhou
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaixin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunyang Pan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haojie Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Gong
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Ishikura T, Kinoshita M, Shimizu M, Yasumizu Y, Motooka D, Okuzaki D, Yamashita K, Murata H, Beppu S, Koda T, Tada S, Shiraishi N, Sugiyama Y, Miyamoto K, Kusunoki S, Sugimoto T, Kumanogoh A, Okuno T, Mochizuki H. Anti-AQP4 autoantibodies promote ATP release from astrocytes and induce mechanical pain in rats. J Neuroinflammation 2021; 18:181. [PMID: 34419102 PMCID: PMC8380350 DOI: 10.1186/s12974-021-02232-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/04/2021] [Indexed: 12/23/2022] Open
Abstract
Background Intractable neuropathic pain is a common symptom of neuromyelitis optica spectrum disorder (NMOSD). However, the underlying mechanism of NMOSD pain remains to be elucidated. In this study, we focused on ATP, which is one of the damage-associated molecular patterns, and also a well-recognized molecule involved in peripheral neuropathic pain. Methods We assessed the development of pain symptoms by injecting anti-AQP4 recombinant autoantibodies (rAQP4 IgG) into rat spinal cords. We incubated HEK293 cells expressing AQP4 (HEK-AQP4) and rat astrocytes with rAQP4 IgG and assessed the level of ATP in the supernatant. We performed transcriptome analysis of the spinal cords injected with rAQP4 IgG. Pharmacological inhibition was also applied to investigate the involvement of ATP in the development of neuropathic pain in our rat model. The ATP concentration within the cerebrospinal fluid was examined in patients with NMOSD and other neurological diseases. Results Development of mechanical allodynia was confirmed in rAQP4 IgG–treated rats. AQP4-Ab–mediated extracellular ATP release from astrocytes was observed in vitro, and pharmacological inhibition of ATP receptor reversed mechanical allodynia in the rAQP4 IgG–treated rats. Furthermore, transcriptome analysis revealed elevation of gene expressions related to several ATP receptors including P2rx4 and IL1B in the spinal cord of rAQP4 IgG–treated rats. In patients, CSF ATP concentration was significantly higher in the acute and remission phase of NMOSD than in multiple sclerosis or other neurological disorders. Conclusion Anti-AQP4 antibody was shown to induce the release of extracellular ATP from astrocytes. The ATP-mediated development of mechanical allodynia was also suggested in rats treated with anti-AQP4 antibody. Our study indicates the pivotal role of ATP in the pain mechanism of NMOSD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02232-w.
Collapse
Affiliation(s)
- Teruyuki Ishikura
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Mikito Shimizu
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiaki Yasumizu
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kazuya Yamashita
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hisashi Murata
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shohei Beppu
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toru Koda
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Tada
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naoyuki Shiraishi
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuko Sugiyama
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Katsuichi Miyamoto
- Department of Neurology, Kindai University Faculty of Medicine, Sayama, Osaka, Japan
| | - Susumu Kusunoki
- Department of Neurology, Kindai University Faculty of Medicine, Sayama, Osaka, Japan
| | - Tomoyuki Sugimoto
- Graduate School of Data Science, Shiga University, Hikone, Shiga, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
12
|
Winkler A, Wrzos C, Haberl M, Weil MT, Gao M, Möbius W, Odoardi F, Thal DR, Chang M, Opdenakker G, Bennett JL, Nessler S, Stadelmann C. Blood-brain barrier resealing in neuromyelitis optica occurs independently of astrocyte regeneration. J Clin Invest 2021; 131:141694. [PMID: 33645550 DOI: 10.1172/jci141694] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/06/2021] [Indexed: 01/19/2023] Open
Abstract
Approximately 80% of neuromyelitis optica spectrum disorder (NMOSD) patients harbor serum anti-aquaporin-4 autoantibodies targeting astrocytes in the CNS. Crucial for NMOSD lesion initiation is disruption of the blood-brain barrier (BBB), which allows the entrance of Abs and serum complement into the CNS and which is a target for new NMOSD therapies. Astrocytes have important functions in BBB maintenance; however, the influence of their loss and the role of immune cell infiltration on BBB permeability in NMOSD have not yet been investigated. Using an experimental model of targeted NMOSD lesions in rats, we demonstrate that astrocyte destruction coincides with a transient disruption of the BBB and a selective loss of occludin from tight junctions. It is noteworthy that BBB integrity is reestablished before astrocytes repopulate. Rather than persistent astrocyte loss, polymorphonuclear leukocytes (PMNs) are the main mediators of BBB disruption, and their depletion preserves BBB integrity and prevents astrocyte loss. Inhibition of PMN chemoattraction, activation, and proteolytic function reduces lesion size. In summary, our data support a crucial role for PMNs in BBB disruption and NMOSD lesion development, rendering their recruitment and activation promising therapeutic targets.
Collapse
Affiliation(s)
| | | | - Michael Haberl
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, Göttingen, Germany
| | - Marie-Theres Weil
- Electron Microscopy Core Unit, Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Ming Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany.,Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Francesca Odoardi
- Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, Göttingen, Germany
| | - Dietmar R Thal
- Department of Imaging and Pathology, KU Leuven, and Department of Pathology, UZ Leuven, Leuven, Belgium.,Laboratory of Neuropathology, Institute of Pathology, Ulm University, Ulm, Germany
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Program in Neuroscience, University of Colorado at Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | |
Collapse
|
13
|
Li W, Liu J, Tan W, Zhou Y. The role and mechanisms of Microglia in Neuromyelitis Optica Spectrum Disorders. Int J Med Sci 2021; 18:3059-3065. [PMID: 34400876 PMCID: PMC8364446 DOI: 10.7150/ijms.61153] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune neurological disease that can cause blindness and disability. As the major mediators in the central nervous system, microglia plays key roles in immunological regulation in neuroinflammatory diseases, including NMOSD. Microglia can be activated by interleukin (IL)-6 and type I interferons (IFN-Is) during NMOSD, leading to signal transducer and activator of transcription (STAT) activation. Moreover, complement C3a secreted from activated astrocytes may induce the secretion of complement C1q, inflammatory cytokines and progranulin (PGRN) by microglia, facilitating injury to microglia, neurons, astrocytes and oligodendrocytes in an autocrine or paracrine manner. These processes involving activated microglia ultimately promote the pathological course of NMOSD. In this review, recent research progress on the roles of microglia in NMOSD pathogenesis is summarized, and the mechanisms of microglial activation and microglial-mediated inflammation, and the potential research prospects associated with microglial activation are also discussed.
Collapse
Affiliation(s)
- Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Jiaqin Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Institute of Clinical Pharmacy, Central South University, Changsha, 410011, Hunan, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| |
Collapse
|
14
|
Timmerman R, Burm SM, Bajramovic JJ. Tissue-specific features of microglial innate immune responses. Neurochem Int 2020; 142:104924. [PMID: 33248205 DOI: 10.1016/j.neuint.2020.104924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
As tissue-resident macrophages of the brain, microglia are increasingly considered as cellular targets for therapeutical intervention. Innate immune responses in particular have been implicated in central nervous system (CNS) infections, neuro-oncology, neuroinflammatory and neurodegenerative diseases. We here review the impact of 'nature and nurture' on microglial innate immune responses and summarize documented tissue-specific adaptations. Overall, such adaptations are associated with regulatory processes rather than with overt differences in the expressed repertoire of activating receptors of different tissue-resident macrophages. Microglial responses are characterized by slower kinetics, by a more persistent nature and by a differential usage of downstream enzymes and accessory receptors. We further consider factors like aging, previous exposure to inflammatory stimuli, and differences in the microenvironment that can modulate innate immune responses. The long-life span of microglia in the metabolically active CNS renders them susceptible to the phenomenon of 'inflammaging', and major challenges lie in the unraveling of the factors that underlie age-related alterations in microglial behavior.
Collapse
Affiliation(s)
- R Timmerman
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - S M Burm
- Genmab, Utrecht, the Netherlands
| | - J J Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Rijswijk, the Netherlands.
| |
Collapse
|
15
|
Shimizu M, Okuno T, Kinoshita M, Sumi H, Fujimura H, Yamashita K, Sugimoto T, Sakakibara S, Sakakibara K, Koda T, Tada S, Ishikura T, Murata H, Beppu S, Shiraishi N, Sugiyama Y, Nakatsuji Y, Kumanogoh A, Mochizuki H. Mitochondrial DNA enhance innate immune responses in neuromyelitis optica by monocyte recruitment and activation. Sci Rep 2020; 10:13274. [PMID: 32764561 PMCID: PMC7414017 DOI: 10.1038/s41598-020-70203-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 07/22/2020] [Indexed: 11/24/2022] Open
Abstract
Although recent studies indicate the involvement of monocytes in accelerating the lesion formation of neuromyelitis optica spectrum disorder (NMOSD), the precise mechanism of the innate immune system activation remains elusive. Thus, in this study, we aimed to clarify the mechanisms of NMOSD pathogenesis from the viewpoint of innate immunity activation. We established anti-AQP4 recombinant autoantibodies (Ab) from plasmablasts in NMOSD patient's CSF. Human astrocytes treated with anti-AQP4 Ab produced a significant amount of CCL2 and contributed to the efficient recruitment of monocytes. Moreover, mitochondrial DNA (mtDNA), which activated monocytes via Toll-like receptor 9 (TLR9), was released from astrocytes treated with anti-AQP4 Ab. MtDNA further enhanced CCL2 production by monocytes, and it was demonstrated that mtDNA concentration correlated with the efficiency of monocyte recruitment in the CSF of NMOSD patients. In conclusion, these observations highlight that mtDNA which was released from astrocytes damaged by anti-AQP4 Ab has a central role in establishing the inflammatory loop of monocyte recruitment and activation via an innate immunity pathway.
Collapse
Affiliation(s)
- Mikito Shimizu
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hisae Sumi
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Harutoshi Fujimura
- Department of Neurology, Osaka-Toneyama National Medical Center, 5-1-1, Toneyama, Toyonaka, Osaka, 560-8552, Japan
| | - Kazuya Yamashita
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomoyuki Sugimoto
- Faculty of Data Science, Shiga University, 1-1-1, Baba, Hikone, Shiga, 522-8522, Japan
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kaori Sakakibara
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toru Koda
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Tada
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Teruyuki Ishikura
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hisashi Murata
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shohei Beppu
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naoyuki Shiraishi
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuko Sugiyama
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuji Nakatsuji
- Department of Neurology, Faculty of Medicine, University of Toyama, 2630, Sugitani, Toyama, 930-0194, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
16
|
Ramakrishnan P, Nagarajan D. Neuromyelitis optica spectrum disorder: an overview. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
17
|
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
|
18
|
West PK, Viengkhou B, Campbell IL, Hofer MJ. Microglia responses to interleukin-6 and type I interferons in neuroinflammatory disease. Glia 2019; 67:1821-1841. [PMID: 31033014 DOI: 10.1002/glia.23634] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2025]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They are a heterogenous, exquisitely responsive, and highly plastic cell population, which enables them to perform diverse roles. They sense and respond to the local production of many different signals, including an assorted range of cytokines. Microglia respond strongly to interleukin-6 (IL-6) and members of the type I interferon (IFN-I) family, IFN-alpha (IFN-α), and IFN-beta (IFN-β). Although these cytokines are essential in maintaining homeostasis and for activating and regulating immune responses, their chronic production has been linked to the development of distinct human neurological diseases, termed "cerebral cytokinopathies." IL-6 and IFN-α have been identified as key mediators in the pathogenesis of neuroinflammatory disorders including neuromyelitis optica and Aicardi-Goutières syndrome, respectively, whereas IFN-β has an emerging role as a causal factor in age-associated cognitive decline. One of the key features that unites these diseases is the presence of highly reactive microglia. The current understanding is that microglia contribute to the development of cerebral cytokinopathies and represent an important therapeutic target. However, it remains to be resolved whether microglia have beneficial or detrimental effects. Here we review and discuss what is currently known about the microglial response to IL-6 and IFN-I, based on both animal models and clinical studies. Foundational knowledge regarding the microglial response to IL-6 and IFN-I is now being used to devise therapeutic strategies to ameliorate neuroinflammation and promote repair: either through targeting microglia, or by targeting the reduction of CNS levels or downstream biological pathways of IL-6 or IFN-I.
Collapse
Affiliation(s)
- Phillip K West
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Barney Viengkhou
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Jin X, Wang T, Liao Y, Guo J, Wang G, Zhao F, Jin Y. Neuroinflammatory Reactions in the Brain of 1,2-DCE-Intoxicated Mice during Brain Edema. Cells 2019; 8:cells8090987. [PMID: 31461951 PMCID: PMC6770564 DOI: 10.3390/cells8090987] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 08/24/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
We previously reported that expression of matrix metalloproteinase-9 (MMP-9) mRNA and protein was upregulated during 1,2-dichloroethane (1,2-DCE) induced brain edema in mice. We also found that the p38 mitogen-activated protein kinase (p38 MAPK) signaling pathway resulted in MMP-9 overexpression and nuclear factor-κB (NF-κB) activation in mice treated with 1,2-DCE. In this study, we further hypothesized that inflammatory reactions mediated by the p38 MAPK/ NF-κB signaling pathway might be involved in MMP-9 overexpression, blood–brain barrier (BBB) disruption and edema formation in the brain of 1,2-DCE-intoxicated mice. Our results revealed that subacute poisoning by 1,2-DCE upregulates protein levels of glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter molecule 1 (Iba-1), interleukin-1β (IL-1β), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), inducible nitric oxide synthase (iNOS) and p-p65 in mouse brains. Pretreatment with an inhibitor against p38 MAPK attenuates these changes. Moreover, pretreatment with an inhibitor against NF-κB attenuates alterations in brain water content, pathological indications notable in brain edema, as well as mRNA and protein expression on levels of MMP-9, VCAM-1, ICAM-1, iNOS, and IL-1β, tight junction proteins (TJs), GFAP and Iba-1 in the brain of 1,2-DCE-intoxicated mice. Furthermore, pretreatment with an inhibitor against MMP-9 obstructs the decrease of TJs in the brain of 1,2-DCE-intoxicated mice. Lastly, pretreatment with an antagonist against the IL-1β receptor also attenuates changes in protein levels of p-p38 MAPK, p-p65, p-IκB, VCAM -1, ICAM-1, IL-1β, and Iba-1 in the brain of 1,2-DCE-intoxicated-mice. Taken together, findings from the current study indicate that the p38 MAPK/ NF-κB signaling pathway might be involved in the activation of glial cells, and the overproduction of proinflammatory factors, which might induce inflammatory reactions in the brain of 1,2-DCE-intoxicated mice that leads to brain edema.
Collapse
Affiliation(s)
- Xiaoxia Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
- Department of Occupational and Environmental Health, School of Public Health, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yingjun Liao
- Department of Physiology, China Medical University, Shenyang 110122, Liaoning, China
| | - Jingjing Guo
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
20
|
Abstract
Purpose of review Neuromyelitis optica spectrum disorders (NMOSD) are severe inflammatory diseases of the central nervous system (CNS), with the presence of aquaporin 4 (AQP4)-specific serum antibodies in the vast majority of patients, and with the presence of myelin oligodendrocyte glycoprotein (MOG)-specific antibodies in approximately 40% of all AQP4-antibody negative NMOSD patients. Despite differences in antigen recognition, the preferred sites of lesions are similar in both groups of patients: They localize to the spinal cord and to the anterior visual pathway including retina, optic nerves, chiasm, and optic tracts, and – to lesser extent – also to certain predilection sites in the brain. Recent findings The involvement of T cells in the formation of NMOSD lesions has been challenged for quite some time. However, several recent findings demonstrate the key role of T cells for lesion formation and localization. Studies on the evolution of lesions in the spinal cord of NMOSD patients revealed a striking similarity of early NMOSD lesions with those observed in corresponding T-cell-induced animal models, both in lesion formation and in lesion localization. Studies on retinal abnormalities in NMOSD patients and corresponding animals revealed the importance of T cells for the very early stages of retinal lesions which eventually culminate in damage to Müller cells and to the retinal nerve fiber layer. Finally, a study on cerebrospinal fluid (CSF) barrier pathology demonstrated that NMOSD immunopathology extends beyond perivascular astrocytic foot processes to include the pia, the ependyma, and the choroid plexus, and that diffusion of antibodies from the CSF could further influence lesion formation in NMOSD patients. Summary The pathological changes observed in AQP4-antibody positive and MOG-antibody positive NMOSD patients are strikingly similar to those found in corresponding animal models, and many mechanisms which determine lesion localization in experimental animals seem to closely reflect the human situation.
Collapse
|
21
|
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
|
22
|
Yamashita K, Kinoshita M, Miyamoto K, Namba A, Shimizu M, Koda T, Sugimoto T, Mori Y, Yoshioka Y, Nakatsuji Y, Kumanogoh A, Kusunoki S, Mochizuki H, Okuno T. Cerebrospinal fluid mitochondrial DNA in neuromyelitis optica spectrum disorder. J Neuroinflammation 2018; 15:125. [PMID: 29703264 PMCID: PMC5924507 DOI: 10.1186/s12974-018-1162-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/17/2018] [Indexed: 12/14/2022] Open
Abstract
Background Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory disease of the central nervous system. Although complement-dependent astrocyte damage mediated by anti-aquaporin 4 autoantibody (AQP4-Ab) is well acknowledged to be the core of NMOSD pathogenesis, additional inflammatory cascades may contribute to the establishment of lesion formation. Thus, in this study, we investigated the possible pathogenic role of immune-reactive mitochondrial DNA (mtDNA) in cerebrospinal fluid (CSF) of NMOSD patients. Methods Using quantitative polymerase chain reaction, we measured extracellular mtDNA levels in CSF of NMOSD patients positive for AQP4-Ab. Patients with multiple sclerosis or other neurological diseases were examined as controls. Pre- and post-treatment extracellular mtDNA levels were also compared in the NMOSD group. Extracellular mtDNA release from human astrocytes was analyzed in vitro utilizing NMOSD sera, and interleukin (IL)-1β production was measured in supernatants of mixed glial cells stimulated with DNA fraction of CSF derived from NMOSD patients. Furthermore, specific innate immune pathways mediating the IL-1β production by mtDNA were investigated in peripheral blood mononuclear cells with selective inhibitors of Toll-like receptor 9 (TLR9) and NOD-like receptor protein 3 (NLRP3) inflammasomes. Results Extracellular mtDNA level was specifically elevated in acute phase of NMOSD CSF. In vitro studies provided the evidence that mtDNA is released from human astrocytes by NMOSD sera. In addition, DNA fraction isolated from NMOSD CSF promoted secretion of IL-1β from mixed glial cells. Selective inhibition of TLR9 and NLRP3 inflammasomes revealed that mtDNA-mediated IL-1β production depends on specific innate immune pathways. Conclusion Extracellular mtDNA is specifically elevated in the CSF of patients with acute phase NMOSD, and mtDNA released by AQP4-Ab-mediated cellular damage elicits the innate immune cascades via TLR9 and NLRP3 inflammasomes pathways. Our study highlights mtDNA-mediated innate immune pathways as a novel therapeutic target for future treatment of NMOSD patients. Electronic supplementary material The online version of this article (10.1186/s12974-018-1162-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kazuya Yamashita
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan
| | - Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan.,Department of Neurology, Osaka General Medical Center, Osaka, Japan
| | - Katsuichi Miyamoto
- Department of Neurology, Kinki University Graduate School of Medicine, Osaka, Japan
| | - Akiko Namba
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan
| | - Mikito Shimizu
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan
| | - Toru Koda
- National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tomoyuki Sugimoto
- Department of Mathematics and Computer Science, Kagoshima University Graduate School of Science and Technology, Kagoshima, Japan
| | - Yuki Mori
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT), Osaka University, Osaka, Japan
| | - Yoshichika Yoshioka
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology (NICT), Osaka University, Osaka, Japan
| | - Yuji Nakatsuji
- Department of Neurology, Toyama University, Toyama, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Susumu Kusunoki
- Department of Neurology, Kinki University Graduate School of Medicine, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan.
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, D4, 2-2 Yamadaoka, Osaka, 565-0871, Japan.
| |
Collapse
|
23
|
Silva BA, Leal MC, Farías MI, Avalos JC, Besada CH, Pitossi FJ, Ferrari CC. A new focal model resembling features of cortical pathology of the progressive forms of multiple sclerosis: Influence of innate immunity. Brain Behav Immun 2018; 69:515-531. [PMID: 29378262 DOI: 10.1016/j.bbi.2018.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/10/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating disease of unknown aetiology that causes neurological disabilities in young adults. MS displays different clinical patterns, including recurrent episodes with remission periods ("relapsing-remitting MS" (RRMS)), which can progress over several years to a secondary progressive form (SPMS). However, 10% of patients display persistent progression at the onset of disease ("primary progressive MS" (PPMS)). Currently, no specific therapeutic agents are available for the progressive forms, mainly because the underlying pathogenic mechanisms are not clear and because no animal models have been specifically developed for these forms. The development of MS animal models is required to clarify the pathological mechanisms and to test novel therapeutic agents. In the present work, we overexpressed interleukin 1 beta (IL-1β) in the cortex to develop an animal model reflecting the main pathological hallmarks of MS. The treated animals presented with neuroinflammation, demyelination, glial activation, and neurodegeneration along with cognitive symptoms and MRI images consistent with MS pathology. We also demonstrated the presence of meningeal inflammation close to cortical lesions, with characteristics similar to those described in MS patients. Systemic pro-inflammatory stimulation caused a flare-up of the cortical lesions and behavioural symptoms, including impairment of working memory and the appearance of anxiety-like symptoms. Our work demonstrated induced cortical lesions, reflecting the main histopathological hallmarks and cognitive impairments characterizing the cortical pathology described in MS patients with progressive forms of the disease.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Celeste Leal
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - María Isabel Farías
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | | | | | - Fernando Juan Pitossi
- Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Institute of Basic Science and Experimental Medicine (ICBME), University Institute, Italian Hospital, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations of Buenos Aires, (IIBBA, CONICET), Buenos Aires, Argentina.
| |
Collapse
|
24
|
Tröscher AR, Klang A, French M, Quemada-Garrido L, Kneissl SM, Bien CG, Pákozdy Á, Bauer J. Selective Limbic Blood-Brain Barrier Breakdown in a Feline Model of Limbic Encephalitis with LGI1 Antibodies. Front Immunol 2017; 8:1364. [PMID: 29093718 PMCID: PMC5651237 DOI: 10.3389/fimmu.2017.01364] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022] Open
Abstract
Human leucine-rich glioma-inactivated protein 1 encephalitis (LGI1) is an autoimmune limbic encephalitis in which serum and cerebrospinal fluid contain antibodies targeting LGI1, a protein of the voltage gated potassium channel (VGKC) complex. Recently, we showed that a feline model of limbic encephalitis with LGI1 antibodies, called feline complex partial seizures with orofacial involvement (FEPSO), is highly comparable to human LGI1 encephalitis. In human LGI1 encephalitis, neuropathological investigations are difficult because very little material is available. Taking advantage of this natural animal model to study pathological mechanisms will, therefore, contribute to a better understanding of its human counterpart. Here, we present a brain-wide histopathological analysis of FEPSO. We discovered that blood–brain barrier (BBB) leakage was present not only in all regions of the hippocampus but also in other limbic structures such as the subiculum, amygdale, and piriform lobe. However, in other regions, such as the cerebellum, no leakage was observed. In addition, this brain-region-specific immunoglobulin leakage was associated with the breakdown of endothelial tight junctions. Brain areas affected by BBB dysfunction also revealed immunoglobulin and complement deposition as well as neuronal cell death. These neuropathological findings were supported by magnetic resonance imaging showing signal and volume increase in the amygdala and the piriform lobe. Importantly, we could show that BBB disturbance in LGI1 encephalitis does not depend on T cell infiltrates, which were present brain-wide. This finding points toward another, so far unknown, mechanism of opening the BBB. The limbic predilection sites of immunoglobulin antibody leakage into the brain may explain why most patients with LGI1 antibodies have a limbic phenotype even though LGI1, the target protein, is ubiquitously distributed across the central nervous system.
Collapse
Affiliation(s)
- Anna R Tröscher
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Andrea Klang
- Department for Pathobiology, Institute of Pathology and Forensic Veterinary Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Maria French
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Lucía Quemada-Garrido
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Sibylle Maria Kneissl
- Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| | | | - Ákos Pákozdy
- Clinical Unit of Internal Medicine Small Animals, University of Veterinary Medicine, Vienna, Austria
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Kong BS, Kim Y, Kim GY, Hyun JW, Kim SH, Jeong A, Kim HJ. Increased frequency of IL-6-producing non-classical monocytes in neuromyelitis optica spectrum disorder. J Neuroinflammation 2017; 14:191. [PMID: 28946890 PMCID: PMC5613387 DOI: 10.1186/s12974-017-0961-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system that preferentially affects the optic nerves, spinal cord, and area postrema. A series of evidence suggested that B cells play a fundamental role in the pathogenesis of NMOSD. However, there are still gaps left to be answered in NMOSD pathogenesis suggesting the roles of other immune cells. This study aimed to investigate the monocyte inflammatory characteristics, monocyte subset frequency and cytokine production, and cell-surface molecule expression in NMOSD, multiple sclerosis (MS), and healthy controls (HC). METHODS Peripheral blood mononuclear cells of 20 aquaporin 4IgG-positive NMOSD patients, 20 MS patients, and 20 healthy controls were collected to analyze the monocyte subsets and to purify monocytes. To mimic the adaptive immunity, we have activated the monocytes using CD40L and IFN-γ to observe the production of cytokines and expression of cell-surface molecules. RESULTS NMOSD monocytes showed a remarkable increase in the production of pro-inflammatory cytokines (IL-6, IL-1β) and increased expression of cell-surface molecules (CD80, HLA, ICAM-1, CD16), as well as a decrease in the levels of anti-inflammatory cytokine IL-10, compared to healthy control (HC) monocytes. As expected, MS monocytes also exhibit increased inflammatory cytokine production and increased cell-surface molecule expression compared to HC monocytes. Further analysis of monocyte subsets revealed that NMOSD monocytes have an increased frequency of the non-classical monocyte subset (CD14+CD16++) and a decreased frequency of the classical monocyte subset (CD14++CD16+) compared to HC monocytes. This finding was distinctly different from that of MS monocytes, which had an increased intermediate monocyte (CD14+CD16+) subset. In addition, these NMOSD non-classical monocyte subsets were highly dedicated, IL-6-producing monocytes. CONCLUSIONS Increased expression of cell-surface molecules and a reciprocal dysregulation of inflammatory and anti-inflammatory cytokines in NMOSD monocytes suggest an altered monocyte inflammatory response. CD14+CD16++ non-classical monocyte subset was more abundant in NMOSD monocytes than in HC or MS monocytes, and NMOSD non-classical monocyte subset had dysregulated IL-6 production, a phenotype which has been reported to be highly associated with NMOSD pathogenesis.
Collapse
Affiliation(s)
- Byung Soo Kong
- Division of Clinical Research, Research Institute and Hospital of the National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, South Korea
| | - Yeseul Kim
- Division of Clinical Research, Research Institute and Hospital of the National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, South Korea
| | - Ga Young Kim
- Division of Clinical Research, Research Institute and Hospital of the National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, South Korea
| | - Jae-Won Hyun
- Department of Neurology, Research Institute and Hospital of the National Cancer Center, Goyang, South Korea
| | - Su-Hyun Kim
- Department of Neurology, Research Institute and Hospital of the National Cancer Center, Goyang, South Korea
| | - Aeran Jeong
- Department of Neurology, Research Institute and Hospital of the National Cancer Center, Goyang, South Korea
| | - Ho Jin Kim
- Division of Clinical Research, Research Institute and Hospital of the National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 10408, South Korea. .,Department of Neurology, Research Institute and Hospital of the National Cancer Center, Goyang, South Korea.
| |
Collapse
|
26
|
Mader S, Brimberg L, Diamond B. The Role of Brain-Reactive Autoantibodies in Brain Pathology and Cognitive Impairment. Front Immunol 2017; 8:1101. [PMID: 28955334 PMCID: PMC5601985 DOI: 10.3389/fimmu.2017.01101] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/22/2017] [Indexed: 12/15/2022] Open
Abstract
Antibodies to different brain proteins have been recently found to be associated with an increasing number of different autoimmune diseases. They need to penetrate the blood–brain barrier (BBB) in order to bind antigens within the central nervous system (CNS). They can target either neuronal or non-neuronal antigen and result in damage either by themselves or in synergy with other inflammatory mediators. Antibodies can lead to acute brain pathology, which may be reversible; alternatively, they may trigger irreversible damage that persists even though the antibodies are no longer present. In this review, we will describe two different autoimmune conditions and the role of their antibodies in causing brain pathology. In systemic lupus erythematosus (SLE), patients can have double stranded DNA antibodies that cross react with the neuronal N-methyl-d-aspartate receptor (NMDAR), which have been recently linked to neurocognitive dysfunction. In neuromyelitis optica (NMO), antibodies to astrocytic aquaporin-4 (AQP4) are diagnostic of disease. There is emerging evidence that pathogenic T cells also play an important role for the disease pathogenesis in NMO since they infiltrate in the CNS. In order to enable appropriate and less invasive treatment for antibody-mediated diseases, we need to understand the mechanisms of antibody-mediated pathology, the acute and chronic effects of antibody exposure, if the antibodies are produced intrathecally or systemically, their target antigen, and what triggers their production. Emerging data also show that in utero exposure to some brain-reactive antibodies, such as those found in SLE, can cause neurodevelopmental impairment since they can penetrate the embryonic BBB. If the antibody exposure occurs at a critical time of development, this can result in irreversible damage of the offspring that persists throughout adulthood.
Collapse
Affiliation(s)
- Simone Mader
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Lior Brimberg
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| | - Betty Diamond
- The Feinstein Institute for Medical Research, The Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, Northwell Health System, Manhasset, NY, United States
| |
Collapse
|
27
|
Efficacy and safety of tacrolimus treatment for neuromyelitis optica spectrum disorder. Sci Rep 2017; 7:831. [PMID: 28400553 PMCID: PMC5429791 DOI: 10.1038/s41598-017-00860-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/15/2017] [Indexed: 12/17/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a severe inflammatory autoimmune disease that mainly involves the optic nerves and spinal cord, causing blindness and paralysis. Although some immunosuppressants such as rituximab and azathioprine have proven to be effective in relapse prevention, the high costs or intolerable adverse events preclude their wide application. Thus, we have conducted a retrospective study in 25 NMOSD patients who were treated with tacrolimus, an immunosuppressant with high efficacy and good tolerance in other autoimmune diseases, to assess its efficacy and safety in NMOSD treatment during the last five years (2011-2016). The results revealed that tacrolimus could reduce the relapse rate by 86.2% and improve the Expanded Disability Status Scale (EDSS) scores (4.5 vs 2.3; P < 0.001) significantly. Relapses in tacrolimus treatment were associated with serum titers of aquaporin 4 antibody (AQP4-IgG) (P = 0.028). Further Cox proportional analysis demonstrated that patients with high titers of AQP4-IgG (≥1:64) had a significantly higher risk of relapse than those with low titers after tacrolimus therapy (HR:5.665; CI95: 1.012-31.705; P = 0.048). Tacrolimus tended to be superior to azathioprine (29 patients) in terms of efficacy and safety during the same period. Our study suggests that tacrolimus may be another promising immunosuppressant for NMOSD.
Collapse
|
28
|
Zeka B, Lassmann H, Bradl M. Müller cells and retinal axons can be primary targets in experimental neuromyelitis optica spectrum disorder. ACTA ACUST UNITED AC 2017; 8:3-7. [PMID: 28344667 PMCID: PMC5347906 DOI: 10.1111/cen3.12345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/21/2016] [Indexed: 01/09/2023]
Abstract
Recent work from our laboratory, using different models of experimental neuromyelitis optica spectrum disorder (NMOSD), has led to a number of observations that might be highly relevant for NMOSD patients. For example: (i) in the presence of neuromyelitis optica immunoglobulin G, astrocyte‐destructive lesions can be initiated by CD4+ T cells when these cells recognize aquaporin 4 (AQP4), but also when they recognize other antigens of the central nervous system. The only important prerequisite is that the T cells have to be activated within the central nervous system by “their” specific antigen. Recently activated CD4+ T cells with yet unknown antigen specificity are also found in human NMOSD lesions. (ii) The normal immune repertoire might contain AQP4‐specific T cells, which are highly encephalitogenic on activation. (iii) The retina might be a primary target of AQP4‐specific T cells and neuromyelitis optica immunoglobulin G: AQP4‐specific T cells alone are sufficient to cause retinitis with low‐grade axonal pathology in the retinal nerve fiber/ganglionic cell layer. A thinning of these layers is also observed in NMOSD patients, where it is thought to be a consequence of optic neuritis. Neuromyelitis optica immunoglobulin G might target cellular processes of Müller cells and cause their loss of AQP4 reactivity, when AQP4‐specific T cells open the blood–retina barrier in the outer plexiform layer. Patchy loss of AQP4 reactivity on Müller cells of NMOSD patients has been recently described. Cumulatively, our findings in experimental NMOSD suggest that both CD4+ T cell and antibody responses directed against AQP4 might play an important role in the pathogenesis of tissue destruction seen in NMOSD.
Collapse
Affiliation(s)
- Bleranda Zeka
- Department for Neuroimmunology Center for Brain Research Medical University Vienna Vienna Austria
| | - Hans Lassmann
- Department for Neuroimmunology Center for Brain Research Medical University Vienna Vienna Austria
| | - Monika Bradl
- Department for Neuroimmunology Center for Brain Research Medical University Vienna Vienna Austria
| |
Collapse
|
29
|
Zeng Q, Dong X, Ruan C, Hu B, Luo Y, Luo Z, Xu L, Zhou H, Wang R, Yang H. CD14 +CD16 ++ monocytes are increased in patients with NMO and are selectively suppressed by glucocorticoids therapy. J Neuroimmunol 2016; 300:1-8. [PMID: 27806868 DOI: 10.1016/j.jneuroim.2016.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 11/30/2022]
Abstract
The pathophysiologic significance of the CD16+ monocyte subset has been demonstrated by its expansion in various autoimmune disorders. To date, the characteristics and roles of monocyte subpopulations in patients with neuromyelitis optica (NMO) have been poorly defined. We measured the percentages of the monocyte subsets in the peripheral blood, the levels of IL-1β and TNF-α mRNA in monocyte subsets and the concentrations of IL-1β and TNF-α in plasma and CSF from NMO patients. Our results showed that nonclassical monocytes were up-regulated in NMO patients and significantly elevated IL-1β and TNF-α expression was detected in it. In addition the increased nonclassical monocytes could be selectively suppressed by GC in patients with NMO.
Collapse
Affiliation(s)
- Qiuming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Xiaohua Dong
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Chunyun Ruan
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Bo Hu
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Yuebei Luo
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Liqun Xu
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Hao Zhou
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Runqi Wang
- Department of Neurology, Xiangya Hospital, Central South University, PR China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, PR China.
| |
Collapse
|
30
|
Burm SM, Peferoen LAN, Zuiderwijk-Sick EA, Haanstra KG, 't Hart BA, van der Valk P, Amor S, Bauer J, Bajramovic JJ. Expression of IL-1β in rhesus EAE and MS lesions is mainly induced in the CNS itself. J Neuroinflammation 2016; 13:138. [PMID: 27266875 PMCID: PMC4895983 DOI: 10.1186/s12974-016-0605-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/30/2016] [Indexed: 12/21/2022] Open
Abstract
Background Interleukin (IL)-1β is a pro-inflammatory cytokine that plays a role in the pathogenesis of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), the animal model for MS. Yet, detailed studies on IL-1β expression in different stages of MS lesion development and a comparison of IL-1β expression in MS and EAE are lacking. Methods Here, we performed an extensive characterization of IL-1β expression in brain tissue of MS patients, which included different MS lesion types, and in brain tissue of rhesus macaques with EAE. Results In rhesus EAE brain tissue, we observed prominent IL-1β staining in MHC class II+ cells within perivascular infiltrates and at the edges of large demyelinating lesions. Surprisingly, staining was localized to resident microglia or differentiated macrophages rather than to infiltrating monocytes, suggesting that IL-1β expression is induced within the central nervous system (CNS). By contrast, IL-1β staining in MS brain tissue was much less pronounced. Staining was found in the parenchyma of active and chronic active MS lesions and in nodules of MHC class II+ microglia in otherwise normal appearing white matter. IL-1β expression was detected in a minority of the nodules only, which could not be distinguished by the expression of pro- and anti-inflammatory markers. These nodules were exclusively found in MS, and it remains to be determined whether IL-1β+ nodules are destined to progress into active lesions or whether they merely reflect a transient response to cellular stress. Conclusions Although the exact localization and relative intensity of IL-1β expression in EAE and MS is different, the staining pattern in both neuroinflammatory disorders is most consistent with the idea that the expression of IL-1β during lesion development is induced in the tissue rather than in the periphery. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0605-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Saskia Maria Burm
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | | | - Ella Alwine Zuiderwijk-Sick
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Krista Geraldine Haanstra
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Bert Adriaan 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - Paul van der Valk
- Department of Pathology, VU Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, VU Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Jan Bauer
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria
| | - Jeffrey John Bajramovic
- Alternatives Unit, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
| |
Collapse
|
31
|
Marignier R, Ruiz A, Cavagna S, Nicole A, Watrin C, Touret M, Parrot S, Malleret G, Peyron C, Benetollo C, Auvergnon N, Vukusic S, Giraudon P. Neuromyelitis optica study model based on chronic infusion of autoantibodies in rat cerebrospinal fluid. J Neuroinflammation 2016; 13:111. [PMID: 27193196 PMCID: PMC4872335 DOI: 10.1186/s12974-016-0577-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/10/2016] [Indexed: 11/19/2022] Open
Abstract
Background Devic’s neuromyelitis optica (NMO) is an autoimmune astrocytopathy, associated with central nervous system inflammation, demyelination, and neuronal injury. Several studies confirmed that autoantibodies directed against aquaporin-4 (AQP4-IgG) are relevant in the pathogenesis of NMO, mainly through complement-dependent toxicity leading to astrocyte death. However, the effect of the autoantibody per se and the exact role of intrathecal AQP4-IgG are still controversial. Methods To explore the intrinsic effect of intrathecal AQP4-IgG, independent from additional inflammatory effector mechanisms, and to evaluate its clinical impact, we developed a new animal model, based on a prolonged infusion of purified immunoglobulins from NMO patient (IgGAQP4+, NMO-rat) and healthy individual as control (Control-rat) in the cerebrospinal fluid (CSF) of live rats. Results We showed that CSF infusion of purified immunoglobulins led to diffusion in the brain, spinal cord, and optic nerves, the targeted structures in NMO. This was associated with astrocyte alteration in NMO-rats characterized by loss of aquaporin-4 expression in the spinal cord and the optic nerves compared to the Control-rats (p = 0.001 and p = 0.02, respectively). In addition, glutamate uptake tested on vigil rats was dramatically reduced in NMO-rats (p = 0.001) suggesting that astrocytopathy occurred in response to AQP4-IgG diffusion. In parallel, myelin was altered, as shown by the decrease of myelin basic protein staining by up to 46 and 22 % in the gray and white matter of the NMO-rats spinal cord, respectively (p = 0.03). Loss of neurofilament positive axons in NMO-rats (p = 0.003) revealed alteration of axonal integrity. Then, we investigated the clinical consequences of such alterations on the motor behavior of the NMO-rats. In a rotarod test, NMO-rats performance was lower compared to the controls (p = 0.0182). AQP4 expression, and myelin and axonal integrity were preserved in AQP4-IgG-depleted condition. We did not find a major immune cell infiltration and microglial activation nor complement deposition in the central nervous system, in our model. Conclusions We establish a link between motor-deficit, NMO-like lesions and astrocytopathy mediated by intrathecal AQP4-IgG. Our study validates the concept of the intrinsic effect of autoantibody against surface antigens and offers a model for testing antibody and astrocyte-targeted therapies in NMO. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0577-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- R Marignier
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France. .,Université Lyon 1, Université de Lyon, Lyon, France. .,Service de Neurologie A, Eugène Devic EDMUS Foundation Against Multiple Sclerosis, Observatoire Français de la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 59 Boulevard Pinel, 69677, Lyon-Bron cedex, France.
| | - A Ruiz
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - S Cavagna
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - A Nicole
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France
| | - C Watrin
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - M Touret
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - S Parrot
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - G Malleret
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - C Peyron
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - C Benetollo
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - N Auvergnon
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| | - S Vukusic
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France.,Service de Neurologie A, Eugène Devic EDMUS Foundation Against Multiple Sclerosis, Observatoire Français de la Sclérose en Plaques, Hôpital Neurologique Pierre Wertheimer, Hospices Civils de Lyon, 59 Boulevard Pinel, 69677, Lyon-Bron cedex, France
| | - P Giraudon
- INSERM U1028, CNRS UMR 5292, Center for Research in Neuroscience of Lyon, Lyon, France.,Université Lyon 1, Université de Lyon, Lyon, France
| |
Collapse
|
32
|
Oji S, Nicolussi EM, Kaufmann N, Zeka B, Schanda K, Fujihara K, Illes Z, Dahle C, Reindl M, Lassmann H, Bradl M. Experimental Neuromyelitis Optica Induces a Type I Interferon Signature in the Spinal Cord. PLoS One 2016; 11:e0151244. [PMID: 26990978 PMCID: PMC4798752 DOI: 10.1371/journal.pone.0151244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/25/2016] [Indexed: 12/15/2022] Open
Abstract
Neuromyelitis optica (NMO) is an acute inflammatory disease of the central nervous system (CNS) which predominantly affects spinal cord and optic nerves. Most patients harbor pathogenic autoantibodies, the so-called NMO-IgGs, which are directed against the water channel aquaporin 4 (AQP4) on astrocytes. When these antibodies gain access to the CNS, they mediate astrocyte destruction by complement-dependent and by antibody-dependent cellular cytotoxicity. In contrast to multiple sclerosis (MS) patients who benefit from therapies involving type I interferons (I-IFN), NMO patients typically do not profit from such treatments. How is I-IFN involved in NMO pathogenesis? To address this question, we made gene expression profiles of spinal cords from Lewis rat models of experimental neuromyelitis optica (ENMO) and experimental autoimmune encephalomyelitis (EAE). We found an upregulation of I-IFN signature genes in EAE spinal cords, and a further upregulation of these genes in ENMO. To learn whether the local I-IFN signature is harmful or beneficial, we induced ENMO by transfer of CNS antigen-specific T cells and NMO-IgG, and treated the animals with I-IFN at the very onset of clinical symptoms, when the blood-brain barrier was open. With this treatment regimen, we could amplify possible effects of the I-IFN induced genes on the transmigration of infiltrating cells through the blood brain barrier, and on lesion formation and expansion, but could avoid effects of I-IFN on the differentiation of pathogenic T and B cells in the lymph nodes. We observed that I-IFN treated ENMO rats had spinal cord lesions with fewer T cells, macrophages/activated microglia and activated neutrophils, and less astrocyte damage than their vehicle treated counterparts, suggesting beneficial effects of I-IFN.
Collapse
Affiliation(s)
- Satoru Oji
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Eva-Maria Nicolussi
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Nathalie Kaufmann
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Bleranda Zeka
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Kathrin Schanda
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Kazuo Fujihara
- Departments of Multiple Sclerosis Therapeutics and Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Zsolt Illes
- Department of Neurology, University of Southern Denmark, Odense, Denmark
| | - Charlotte Dahle
- Department of Clinical Immunology and Transfusion Medicine and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
33
|
Bradl M, Lassmann H. Neurologic autoimmunity: mechanisms revealed by animal models. HANDBOOK OF CLINICAL NEUROLOGY 2016; 133:121-43. [PMID: 27112675 DOI: 10.1016/b978-0-444-63432-0.00008-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Over the last decade, neurologic autoimmunity has become a major consideration in the diagnosis and management of patients with many neurologic presentations. The nature of the associated antibodies and their targets has led to appreciation of the importance of the accessibility of the target antigen to antibodies, and a partial understanding of the different mechanisms that can follow antibody binding. This chapter will first describe the basic principles of autoimmune inflammation and tissue damage in the central and peripheral nervous system, and will then demonstrate what has been learnt about neurologic autoimmunity from circumstantial clinical evidence and from passive, active, and occasionally spontaneous or genetic animal models. It will cover neurologic autoimmune diseases ranging from disorders of neuromuscular transmission, peripheral and ganglionic neuropathy, to diseases of the central nervous system, where autoantibodies are either pathogenic and cause destruction or changes in function of their targets, where they are harmless bystanders of T-cell-mediated tissue damage, or are not involved at all. Finally, this chapter will summarize the relevance of current animal models for studying the different neurologic autoimmune diseases, and it will identify aspects where future animal models need to be improved to better reflect the disease reality experienced by affected patients, e.g., the chronicity or the relapsing/remitting nature of their disease.
Collapse
Affiliation(s)
- Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria.
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
34
|
Kurosawa K, Misu T, Takai Y, Sato DK, Takahashi T, Abe Y, Iwanari H, Ogawa R, Nakashima I, Fujihara K, Hamakubo T, Yasui M, Aoki M. Severely exacerbated neuromyelitis optica rat model with extensive astrocytopathy by high affinity anti-aquaporin-4 monoclonal antibody. Acta Neuropathol Commun 2015; 3:82. [PMID: 26637322 PMCID: PMC4670539 DOI: 10.1186/s40478-015-0259-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/20/2015] [Indexed: 12/22/2022] Open
Abstract
Introduction Neuromyelitis optica (NMO), an autoimmune astrocytopathic disease associated with anti-aquaporin-4 (AQP4) antibody, is characterized by extensive necrotic lesions preferentially involving the optic nerves and spinal cord. However, previous in-vivo experimental models injecting human anti-AQP4 antibodies only resulted in mild spinal cord lesions compared to NMO autopsied cases. Here, we investigated whether the formation of severe NMO-like lesions occurs in Lewis rats in the context of experimental autoimmune encephalomyelitis (EAE), intraperitoneally injecting incremental doses of purified human immunoglobulin-G from a NMO patient (hIgGNMO) or a high affinity anti-AQP4 monoclonal antibody (E5415A), recognizing extracellular domain of AQP4 made by baculovirus display method. Results NMO-like lesions were observed in the spinal cord, brainstem, and optic chiasm of EAE-rats with injection of pathogenic IgG (hIgGNMO and E5415A), but not in control EAE. Only in higher dose E5415A rats, there were acute and significantly severer clinical exacerbations (tetraparesis or moribund) compared with controls, within half day after the injection of pathogenic IgG. Loss of AQP4 was observed both in EAE rats receiving hIgGNMO and E5415A in a dose dependent manner, but the ratio of AQP4 loss in spinal sections became significantly larger in those receiving high dose E5415A up to about 50 % than those receiving low-dose E5415A or hIgGNMO less than 3 %. These lesions were also characterized by extensive loss of glial fibrillary acidic protein but relatively preserved myelin sheaths with perivascular deposition of IgG and C5b-9, which is compatible with post mortem NMO pathology. In high dose E5415A rats, massive neutrophil infiltration was observed especially at the lesion edge, and such lesions were highly vacuolated with partial demyelination and axonal damage. In contrast, such changes were absent in EAE rats receiving low-dose E5415A and hIgGNMO. Conclusions In the present study, we established a severe experimental NMO rat model with highly clinical exacerbation and extensive tissue destructive lesions typically observed in NMO patients, which has not adequately been realized in in-vivo rodent models. Our data suggest that the pathogenic antibodies could induce immune mediated astrocytopathy with mobilized neutrophils, resulted in early lesion expansion of NMO lesion with vacuolation and other tissue damages. (350/350) Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0259-2) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Zeka B, Hastermann M, Hochmeister S, Kögl N, Kaufmann N, Schanda K, Mader S, Misu T, Rommer P, Fujihara K, Illes Z, Leutmezer F, Sato DK, Nakashima I, Reindl M, Lassmann H, Bradl M. Highly encephalitogenic aquaporin 4-specific T cells and NMO-IgG jointly orchestrate lesion location and tissue damage in the CNS. Acta Neuropathol 2015; 130:783-98. [PMID: 26530185 PMCID: PMC4654751 DOI: 10.1007/s00401-015-1501-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 01/09/2023]
Abstract
In neuromyelitis optica (NMO), astrocytes become targets for pathogenic aquaporin 4 (AQP4)-specific antibodies which gain access to the central nervous system (CNS) in the course of inflammatory processes. Since these antibodies belong to a T cell-dependent subgroup of immunoglobulins, and since NMO lesions contain activated CD4+ T cells, the question arose whether AQP4-specific T cells might not only provide T cell help for antibody production, but also play an important role in the induction of NMO lesions. We show here that highly pathogenic, AQP4-peptide-specific T cells exist in Lewis rats, which recognize AQP4268–285 as their specific antigen and cause severe panencephalitis. These T cells are re-activated behind the blood–brain barrier and deeply infiltrate the CNS parenchyma of the optic nerves, the brain, and the spinal cord, while T cells with other AQP4-peptide specificities are essentially confined to the meninges. Although AQP4268–285-specific T cells are found throughout the entire neuraxis, they have NMO-typical “hotspots” for infiltration, i.e. periventricular and periaqueductal regions, hypothalamus, medulla, the dorsal horns of spinal cord, and the optic nerves. Most remarkably, together with NMO-IgG, they initiate large astrocyte-destructive lesions which are located predominantly in spinal cord gray matter. We conclude that the processing of AQP4 by antigen presenting cells in Lewis rats produces a highly encephalitogenic AQP4 epitope (AQP4268–285), that T cells specific for this epitope are found in the immune repertoire of normal Lewis rats and can be readily expanded, and that AQP4268–285-specific T cells produce NMO-like lesions in the presence of NMO-IgG.
Collapse
Affiliation(s)
- Bleranda Zeka
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Maria Hastermann
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Sonja Hochmeister
- Department for Neurology, Medical University Graz, Auenbruggerplatz 22, 8036, Graz, Austria
| | - Nikolaus Kögl
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Nathalie Kaufmann
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Kathrin Schanda
- Clinical Department for Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Simone Mader
- Clinical Department for Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, 980-8574, Japan
| | - Paulus Rommer
- University Hospital for Neurology, Medical University Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Kazuo Fujihara
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, 980-8574, Japan
| | - Zsolt Illes
- Department of Neurology, University of Southern Denmark, Sdr Boulevard 29, Odense, 5000, Denmark
| | - Fritz Leutmezer
- University Hospital for Neurology, Medical University Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Douglas Kazutoshi Sato
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, 980-8574, Japan
- Department of Neurology, Faculty of Medicine, University of Sao Paulo, Av. Dr. Arnaldo, 455-4th floor (sl 4110), 01246-903, São Paulo, Brazil
| | - Ichiro Nakashima
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryomachi, Aobaku, Sendai, 980-8574, Japan
| | - Markus Reindl
- Clinical Department for Neurology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Hans Lassmann
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Monika Bradl
- Department for Neuroimmunology, Center for Brain Research, Medical University Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
36
|
Antibodies as Mediators of Brain Pathology. Trends Immunol 2015; 36:709-724. [PMID: 26494046 DOI: 10.1016/j.it.2015.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/17/2015] [Accepted: 09/17/2015] [Indexed: 01/04/2023]
Abstract
The brain is normally sequestered from antibody exposure by the blood brain barrier. However, antibodies can access the brain during fetal development before the barrier achieves full integrity, and in disease states when barrier integrity is compromised. Recent studies suggest that antibodies contribute to brain pathology associated with autoimmune diseases such as systemic lupus erythematosus and neuromyelitis optica, and can lead to transient or permanent behavioral or cognitive abnormalities. We review these findings here and examine the circumstances associated with antibody entry into the brain, the routes of access and the mechanisms that then effect pathology. Understanding these processes and the nature and specificity of neuronal autoantibodies may reveal therapeutic strategies toward alleviating or preventing the neurological pathologies and behavioral abnormalities associated with autoimmune disease.
Collapse
|
37
|
Khorooshi R, Asgari N, Mørch MT, Berg CT, Owens T. Hypersensitivity Responses in the Central Nervous System. Front Immunol 2015; 6:517. [PMID: 26500654 PMCID: PMC4595775 DOI: 10.3389/fimmu.2015.00517] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/22/2015] [Indexed: 12/29/2022] Open
Abstract
Immune-mediated tissue damage or hypersensitivity can be mediated by autospecific IgG antibodies. Pathology results from activation of complement, and antibody-dependent cellular cytotoxicity, mediated by inflammatory effector leukocytes include macrophages, natural killer cells, and granulocytes. Antibodies and complement have been associated to demyelinating pathology in multiple sclerosis (MS) lesions, where macrophages predominate among infiltrating myeloid cells. Serum-derived autoantibodies with predominant specificity for the astrocyte water channel aquaporin-4 (AQP4) are implicated as inducers of pathology in neuromyelitis optica (NMO), a central nervous system (CNS) demyelinating disease where activated neutrophils infiltrate, unlike in MS. The most widely used model for MS, experimental autoimmune encephalomyelitis, is an autoantigen-immunized disease that can be transferred to naive animals with CD4+ T cells, but not with antibodies. By contrast, NMO-like astrocyte and myelin pathology can be transferred to mice with AQP4–IgG from NMO patients. This is dependent on complement, and does not require T cells. Consistent with clinical observations that interferon-beta is ineffective as a therapy for NMO, NMO-like pathology is significantly reduced in mice lacking the Type I IFN receptor. In MS, there is evidence for intrathecal synthesis of antibodies as well as blood–brain barrier (BBB) breakdown, whereas in NMO, IgG accesses the CNS from blood. Transfer models involve either direct injection of antibody and complement to the CNS, or experimental manipulations to induce BBB breakdown. We here review studies in MS and NMO that elucidate roles for IgG and complement in the induction of BBB breakdown, astrocytopathy, and demyelinating pathology. These studies point to significance of T-independent effector mechanisms in neuroinflammation.
Collapse
Affiliation(s)
- Reza Khorooshi
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Nasrin Asgari
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark ; Department of Neurology, Vejle Hospital , Vejle , Denmark
| | - Marlene Thorsen Mørch
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Carsten Tue Berg
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark , Odense , Denmark
| |
Collapse
|
38
|
NFκB signaling drives pro-granulocytic astroglial responses to neuromyelitis optica patient IgG. J Neuroinflammation 2015; 12:185. [PMID: 26423139 PMCID: PMC4590277 DOI: 10.1186/s12974-015-0403-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/22/2015] [Indexed: 01/21/2023] Open
Abstract
Background Astrocytes expressing the aquaporin-4 water channel are a primary target of pathogenic, disease-specific immunoglobulins (IgG) found in patients with neuromyelitis optica (NMO). Immunopathological analyses of active NMO lesions highlight a unique inflammatory phenotype marked by infiltration of granulocytes. Previous studies characterized this granulocytic infiltrate as a response to vasculocentric complement activation and localized tissue destruction. In contrast, we observe that granulocytic infiltration in NMO lesions occurs independently of complement-mediated tissue destruction or active demyelination. These immunopathological findings led to the hypothesis that NMO IgG stimulates astrocyte signaling that is responsible for granulocytic recruitment in NMO. Methods Histopathology was performed on archival formalin-fixed paraffin-embedded autopsy-derived CNS tissue from 23 patients clinically and pathologically diagnosed with NMO or NMO spectrum disorder. Primary murine astroglial cultures were stimulated with IgG isolated from NMO patients or control IgG from healthy donors. Transcriptional responses were assessed by microarray, and translational responses were measured by ELISA. Signaling through the NFκB pathway was measured by western blotting and immunostaining. Results Stimulation of primary murine astroglial cultures with NMO IgG elicited a reactive and inflammatory transcriptional response that involved signaling through the canonical NFκB pathway. This signaling resulted in the release of pro-granulocytic chemokines and was inhibited by the clinically relevant proteasome inhibitors bortezomib and PR-957. Conclusions We propose that the astrocytic NFκB-dependent inflammatory response to stimulation by NMO IgG represents one of the earliest events in NMO pathogenesis, providing a target for therapeutic intervention upstream of irreversible cell death and tissue damage. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0403-8) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Pringproa K, Rungsiwiwut R, Tantilertcharoen R, Praphet R, Pruksananonda K, Baumgärtner W, Thanawongnuwech R. Tropism and Induction of Cytokines in Human Embryonic-Stem Cells-Derived Neural Progenitors upon Inoculation with Highly- Pathogenic Avian H5N1 Influenza Virus. PLoS One 2015; 10:e0135850. [PMID: 26274828 PMCID: PMC4537284 DOI: 10.1371/journal.pone.0135850] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/27/2015] [Indexed: 12/22/2022] Open
Abstract
Central nervous system (CNS) dysfunction caused by neurovirulent influenza viruses is a dreaded complication of infection, and may play a role in some neurodegenerative conditions, such as Parkinson-like diseases and encephalitis lethargica. Although CNS infection by highly pathogenic H5N1 virus has been demonstrated, it is unknown whether H5N1 infects neural progenitor cells, nor whether such infection plays a role in the neuroinflammation and neurodegeneration. To pursue this question, we infected human neural progenitor cells (hNPCs) differentiated from human embryonic stem cells in vitro with H5N1 virus, and studied the resulting cytopathology, cytokine expression, and genes involved in the differentiation. Human embryonic stem cells (BG01) were maintained and differentiated into the neural progenitors, and then infected by H5N1 virus (A/Chicken/Thailand/CUK2/04) at a multiplicity of infection of 1. At 6, 24, 48, and 72 hours post-infection (hpi), cytopathic effects were observed. Then cells were characterized by immunofluorescence and electron microscopy, supernatants quantified for virus titers, and sampled cells studied for candidate genes.The hNPCs were susceptible to H5N1 virus infection as determined by morphological observation and immunofluorescence. The infection was characterized by a significant up-regulation of TNF-α gene expression, while expressions of IFN-α2, IFN-β1, IFN-γ and IL-6 remained unchanged compared to mock-infected controls. Moreover, H5N1 infection did not appear to alter expression of neuronal and astrocytic markers of hNPCs, such as β-III tubulin and GFAP, respectively. The results indicate that hNPCs support H5N1 virus infection and may play a role in the neuroinflammation during acute viral encephalitis.
Collapse
Affiliation(s)
- Kidsadagon Pringproa
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| | - Ruttachuk Rungsiwiwut
- Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Rachod Tantilertcharoen
- Veterinary Diagnostic Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Reunkeaw Praphet
- Institute Product Quality and Standardization, Maejo University, Chiang Mai, Thailand
| | - Kamthorn Pruksananonda
- Human Embryonic Stem Cell Research Center, Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Roongroje Thanawongnuwech
- Department of Pathology, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
40
|
Li M, Yan Y. Experimental models of neuromyelitis optica: current status, challenges and future directions. Neurosci Bull 2015; 31:735-44. [PMID: 26109280 DOI: 10.1007/s12264-015-1552-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022] Open
Abstract
Neuromyelitis optica (NMO) is a recurrent inflammatory disease that predominantly attacks the opticnerves and spinal cord. NMO-IgG, the specific autoantibody present in the vast majority of NMO patients, targets the astrocytic water channel protein aquaporin 4 (AQP4), and differentiates NMO from multiple sclerosis. The growing clinical and research interest in NMO makes it urgent to produce an animal model of NMO. The pathogenic effect of anti-AQP4 antibodies derived from the serum of patients paves the way to generating an experimental model based on the anti-AQP4-mediated astrocyte damage. In this review, we discuss the contribution of experimental models to the understanding of the pathogenesis of the disease and drug development. Key questions raised by the existing models are also discussed.
Collapse
Affiliation(s)
- Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, 85013, USA
| | - Yaping Yan
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
41
|
Markedly increased IP-10 production by blood-brain barrier in neuromyelitis optica. PLoS One 2015; 10:e0122000. [PMID: 25811465 PMCID: PMC4374747 DOI: 10.1371/journal.pone.0122000] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/05/2015] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Severe damage to the blood-brain barrier (BBB) allows anti-aquaporin 4 (AQP4) antibodies to access the astrocytic endfeet in neuromyelitis optica (NMO). In the current study, we identified the pathogenic cytokines/chemokines that are responsible for the BBB malfunction induced by NMO sera. METHODS We measured the levels of 27 cytokines/chemokines in human brain microvascular endothelial cells (BMECs) after exposure to sera obtained from patients with the acute and stable phases of anti-AQP4 antibody-positive NMO spectrum disorder (NMOSD), multiple sclerosis (MS) patients and healthy controls (HC) using a multiplexed fluorescent bead-based immunoassay system. RESULTS The induced protein (IP)-10 level in the cells was markedly increased following exposure to acute phase NMOSD sera. Other cytokines/chemokines including interleukin (IL)-6 and monocyte chemotactic protein (MCP)-1 were also significantly increased in the acute NMOSD group compared to both the MS and HC groups. The up-regulation of the IP-10 levels in the cells after exposure to the acute-phase NMOSD sera was also observed using another specified ELISA, and this effect was significantly decreased during the remission phase in the individual NMOSD patients. Furthermore, the increase in the level of IP-10 after exposure to the sera was significantly correlated with the cerebrospinal fluid/serum albumin ratio. CONCLUSIONS Sera from the acute phase of NMO markedly increased the autocrine secretion of IP-10 by BMECs. The over-production of IP-10 in BMECs may play an important role in the pathogenesis of NMO and may therefore help to mediate the trafficking of T cells expressing its receptor across the BBB.
Collapse
|
42
|
Zajda M, Kaźmierczak B, Zakrzewska-Pniewska B. Aquaporin-4: A novel diagnostic biomarker for seronegative neuromyelitis optica. Mult Scler 2015; 22:125-6. [PMID: 25732841 DOI: 10.1177/1352458515575173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Małgorzata Zajda
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Poland
| | - Beata Kaźmierczak
- Department of Biochemistry, Medical University of Warsaw, Poland Neurodegenerative Diseases Research Group, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
43
|
Abstract
Neuromyelitis optica (NMO) is an autoimmune disorder of the central nervous system directed against astrocytes. Initially diagnosed in individuals with monophasic or relapsing optic neuritis and transverse myelitis, NMO is now recognized as a demyelinating disorder with pleiotropic presentations due to the identification of a specific autoantibody response against the astrocyte water channel aquaporin-4 in the majority of individuals. As visual impairment and neurologic dysfunction in NMO are commonly severe, aggressive treatment of relapses and prophylactic immunomodulatory therapy are the focus of treatment. Although there are no approved treatments for NMO, medications and therapeutic interventions for acute and chronic treatment have been the subject of retrospective study and case reports. The goal of this review is to familiarize the reader with biologic and clinical data supporting current treatments in NMO and highlight future strategies based on advancements in our understanding of NMO pathogenesis.
Collapse
|
44
|
Bradl M, Lassmann H. Experimental models of neuromyelitis optica. Brain Pathol 2014; 24:74-82. [PMID: 24345221 PMCID: PMC4065348 DOI: 10.1111/bpa.12098] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 04/11/2013] [Indexed: 12/11/2022] Open
Abstract
For a long time, the most important inflammatory demyelinating diseases of the central nervous system (CNS), for example, multiple sclerosis (MS) and neuromyelitis optica (NMO), were extremely hard to differentiate, often with severe consequences for affected patients. This changed with the discovery of NMO‐immunoglobulin G (IgG), a specific autoantibody which was detected in the vast majority of NMO patients, and with the demonstration that this autoantibody targets aquaporin 4 (AQP4), a water channel found on astrocytes in the CNS. These findings paved the way for the generation of experimental models of NMO. This chapter will discuss the contribution of experimental models to NMO research and what key questions remain to be addressed.
Collapse
Affiliation(s)
- Monika Bradl
- Department Neuroimmunology, Center for Brain Research, Medical University Vienna, Vienna, Austria
| | | |
Collapse
|
45
|
Lucchinetti CF, Guo Y, Popescu BFG, Fujihara K, Itoyama Y, Misu T. The pathology of an autoimmune astrocytopathy: lessons learned from neuromyelitis optica. Brain Pathol 2014; 24:83-97. [PMID: 24345222 DOI: 10.1111/bpa.12099] [Citation(s) in RCA: 281] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 12/13/2022] Open
Abstract
Neuromyelitis optica (NMO) is a disabling autoimmune astrocytopathy characterized by typically severe and recurrent attacks of optic neuritis and longitudinally extensive myelitis. Until recently, NMO was considered an acute aggressive variant of multiple sclerosis (MS), despite the fact that early studies postulated that NMO and MS may be two distinct diseases with a common clinical picture. With the discovery of a highly specific serum autoantibody (NMO-IgG), Lennon and colleagues provided the first unequivocal evidence distinguishing NMO from MS and other central nervous system (CNS) inflammatory demyelinating disorders. The target antigen of NMO-IgG was confirmed to be aquaporin-4 (AQP4), the most abundant water channel protein in the CNS, mainly expressed on astrocytic foot processes at the blood-brain barrier, subpial and subependymal regions. Pathological studies demonstrated that astrocytes were selectively targeted in NMO as evidenced by the extensive loss of immunoreactivities for the astrocytic proteins, AQP4 and glial fibrillary acidic protein (GFAP), as well as perivascular deposition of immunoglobulins and activation of complement even within lesions with a relative preservation of myelin. In support of these pathological findings, GFAP levels in the cerebrospinal fluid (CSF) during acute NMO exacerbations were found to be remarkably elevated in contrast to MS where CSF-GFAP levels did not substantially differ from controls. Additionally, recent experimental studies showed that AQP4 antibody is pathogenic, resulting in selective astrocyte destruction and dysfunction in vitro, ex vivo and in vivo. These findings strongly suggest that NMO is an autoimmune astrocytopathy where damage to astrocytes exceeds both myelin and neuronal damage. This chapter will review recent neuropathological studies that have provided novel insights into the pathogenic mechanisms, cellular targets, as well as the spectrum of tissue damage in NMO.
Collapse
|
46
|
|
47
|
Aubé B, Lévesque SA, Paré A, Chamma É, Kébir H, Gorina R, Lécuyer MA, Alvarez JI, De Koninck Y, Engelhardt B, Prat A, Côté D, Lacroix S. Neutrophils mediate blood-spinal cord barrier disruption in demyelinating neuroinflammatory diseases. THE JOURNAL OF IMMUNOLOGY 2014; 193:2438-54. [PMID: 25049355 DOI: 10.4049/jimmunol.1400401] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Disruption of the blood-brain and blood-spinal cord barriers (BBB and BSCB, respectively) and immune cell infiltration are early pathophysiological hallmarks of multiple sclerosis (MS), its animal model experimental autoimmune encephalomyelitis (EAE), and neuromyelitis optica (NMO). However, their contribution to disease initiation and development remains unclear. In this study, we induced EAE in lys-eGFP-ki mice and performed single, nonterminal intravital imaging to investigate BSCB permeability simultaneously with the kinetics of GFP(+) myeloid cell infiltration. We observed a loss in BSCB integrity within a day of disease onset, which paralleled the infiltration of GFP(+) cells into the CNS and lasted for ∼4 d. Neutrophils accounted for a significant proportion of the circulating and CNS-infiltrating myeloid cells during the preclinical phase of EAE, and their depletion delayed the onset and reduced the severity of EAE while maintaining BSCB integrity. We also show that neutrophils collected from the blood or bone marrow of EAE mice transmigrate more efficiently than do neutrophils of naive animals in a BBB cell culture model. Moreover, using intravital videomicroscopy, we demonstrate that the IL-1R type 1 governs the firm adhesion of neutrophils to the inflamed spinal cord vasculature. Finally, immunostaining of postmortem CNS material obtained from an acutely ill multiple sclerosis patient and two neuromyelitis optica patients revealed instances of infiltrated neutrophils associated with regions of BBB or BSCB leakage. Taken together, our data provide evidence that neutrophils are involved in the initial events that take place during EAE and that they are intimately linked with the status of the BBB/BSCB.
Collapse
Affiliation(s)
- Benoit Aubé
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada; Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Sébastien A Lévesque
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Alexandre Paré
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Émilie Chamma
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Hania Kébir
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Roser Gorina
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Marc-André Lécuyer
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Jorge I Alvarez
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Yves De Koninck
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Alexandre Prat
- Unité de Neuroimmunologie, Centre d'Excellence en Neuromique, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Médecine, Université de Montréal, Montréal, Quebec H3C 3J7, Canada; and
| | - Daniel Côté
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Québec, Université Laval, Quebec, Quebec G1J 2G3, Canada; Centre d'Optique, Photonique et Laser, Université Laval, Quebec, Quebec G1V 0A6, Canada
| | - Steve Lacroix
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Centre Hospitalier de l'Université Laval, Quebec, Quebec G1V 4G2, Canada; Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, Quebec G1V 0A6, Canada;
| |
Collapse
|
48
|
Sato DK, Callegaro D, de Haidar Jorge FM, Nakashima I, Nishiyama S, Takahashi T, Simm RF, Apostolos-Pereira SL, Misu T, Steinman L, Aoki M, Fujihara K. Cerebrospinal fluid aquaporin-4 antibody levels in neuromyelitis optica attacks. Ann Neurol 2014; 76:305-9. [PMID: 24977390 PMCID: PMC4173125 DOI: 10.1002/ana.24208] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/27/2014] [Accepted: 06/23/2014] [Indexed: 11/16/2022]
Abstract
To elucidate immunopathogenetic roles of aquaporin-4 antibodies in the cerebrospinal fluid (CSF) of neuromyelitis optica spectrum disorders (NMOSD), we analyzed aquaporin-4 antibody titers, cellular and inflammatory markers in the CSF collected from 11 aquaporin-4 antibody seropositive patients. The CSF aquaporin-4 antibody levels during attacks (but not in sera) closely correlated with pleocytosis, inflammatory cytokines including interleukin-6 that can regulate antibody-producing plasmablasts, and glial fibrillary acidic protein levels in the CSF. The amount of aquaporin-4 antibodies present in the central nervous system may have therapeutic implications, as it is associated with astrocyte injury and inflammatory responses during NMOSD attacks. Ann Neurol 2014;76:305–309
Collapse
|
49
|
Krumbholz M, Meinl E. B cells in MS and NMO: pathogenesis and therapy. Semin Immunopathol 2014; 36:339-50. [PMID: 24832354 DOI: 10.1007/s00281-014-0424-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/01/2014] [Indexed: 12/28/2022]
Abstract
B linage cells are versatile players in multiple sclerosis (MS) and neuromyelitis optica/neuromyelitis optica spectrum disorder (NMO). New potential targets of autoantibodies have been described recently. Pathogenic mechanisms extend further to antigen presentation and cytokine production, which are increasingly recognized as therapeutic targets. In addition to pro-inflammatory effects of B cells, they may act also as anti-inflammatory via production of interleukin (IL)-10, IL-35, and other mechanisms. Definition of regulatory B cell subsets is an ongoing issue. Recent studies have provided evidence for a loss of B cell self-tolerance in MS. An immunogenetic approach demonstrated exchange of B cell clones between CSF and blood. The central nervous system (CNS) of MS patients fosters B cell survival, at least partly via BAFF and APRIL. The unexpected increase of relapses in a trial with a soluble BAFF/APRIL receptor (atacicept) suggests that this system is involved in MS, but with features that are not yet understood. In this review, we further discuss evidence for B cell and Ig contribution to human MS and NMO pathogenesis, pro-inflammatory and regulatory B cell effector functions, impaired B cell immune tolerance, the B cell-fostering microenvironment in the CNS, and B cell-targeted therapeutic interventions for MS and NMO, including CD20 depletion (rituximab, ocrelizumab, and ofatumumab), anti-IL6-R (tocilizumab), complement-blocking (eculizumab), inhibitors of AQP4-Ig binding (aquaporumab, small molecular compounds), and BAFF/BAFF-R-targeting agents.
Collapse
Affiliation(s)
- Markus Krumbholz
- Institute of Clinical Neuroimmunology, Ludwig Maximilian University of Munich, Max-Lebsche-Platz 31, 81377, Munich, Germany,
| | | |
Collapse
|
50
|
Asavapanumas N, Verkman AS. Neuromyelitis optica pathology in rats following intraperitoneal injection of NMO-IgG and intracerebral needle injury. Acta Neuropathol Commun 2014; 2:48. [PMID: 24758159 PMCID: PMC4234989 DOI: 10.1186/2051-5960-2-48] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/17/2014] [Indexed: 12/20/2022] Open
Abstract
Introduction Animal models of neuromyelitis optica (NMO) are needed for drug testing and evaluation of NMO disease pathogenesis mechanisms. Results We describe a novel passive-transfer model of NMO in which rats made seropositive for human anti-aquaporin-4 (AQP4) immunoglobulin G antibody (NMO-IgG) by intraperitoneal (IP) injections were subject to intracerebral needle injury. Following a single IP injection, NMO-IgG distributed rapidly to peripheral AQP4-expressing cells (kidney collecting duct, gastric glands, airways, skeletal muscle) and area postrema in brain, but not elsewhere in the central nervous system; however, no pathology was seen in brain, spinal cord, optic nerve or peripheral tissues. After testing various maneuvers to produce NMO-IgG-dependent pathology in brain, we found that transient puncture of brain parenchyma with a 28-gauge needle in NMO-IgG seropositive rats produced robust NMO pathology around the needle track, with loss of AQP4 and glial fibrillary acidic protein, granulocyte and macrophage infiltration, centrovascular deposition of activated complement, and blood–brain barrier disruption, with demyelination by 5 days. Pathology was not seen in rats receiving control (non-NMO) human IgG or in NMO-IgG-seropositive rats made complement-deficient by cobra venom factor. Interestingly, at 1 day a reversible, multifocal astrocytopathy was seen with loss of AQP4 and GFAP (but not myelin) in areas away from the needle track. Conclusions NMO-IgG-seropositivity alone is not sufficient to cause NMO pathology in rats, but a single intracerebral needle insertion, without pre-existing inflammation or infusion of pro-inflammatory factors, was sufficient to produce robust NMO pathology in seropositive rats.
Collapse
|