1
|
He Y, Guo K, Xin J. Complement updates in optic neuritis. Front Neurol 2025; 16:1566771. [PMID: 40206291 PMCID: PMC11978624 DOI: 10.3389/fneur.2025.1566771] [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: 01/25/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Optic neuritis (ON) is an inflammatory condition of the optic nerve associated with demyelinating diseases like multiple sclerosis, neuromyelitis optica spectrum disorder, and myelin oligodendrocyte glycoprotein antibody-associated disease. The complement system is crucial in ON pathogenesis, driving blood-optic nerve barrier disruption, inflammation, and tissue damage. This review explores the complement activation pathways-classical, alternative, and lectin-and their roles in ON progression. Key proteins such as C3, C5, and terminal pathway components are highlighted as central to disease mechanisms. Recent advances in complement-targeted therapies, including C1q blockers, C3 and C5 inhibitors, show promising results in clinical and preclinical studies. Novel therapies, like anaphylatoxin receptor blockers and recombinant factor H, expand the treatment landscape, while plasma exchange remains vital for severe, corticosteroid-resistant cases. Challenges remain, such as ON heterogeneity, the long-term safety of complement inhibition, and the need for personalized approaches. Future studies should focus on unraveling complement-mediated mechanisms, identifying biomarkers, and refining therapeutic strategies. This review highlights the critical role of complement in ON and the latest therapeutic advances to improve patient outcomes.
Collapse
Affiliation(s)
- Yuhong He
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Kai Guo
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Jifu Xin
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
2
|
Segal Y, Soltys J, Clarkson BDS, Howe CL, Irani SR, Pittock SJ. Toward curing neurological autoimmune disorders: Biomarkers, immunological mechanisms, and therapeutic targets. Neuron 2025; 113:345-379. [PMID: 39809275 DOI: 10.1016/j.neuron.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/26/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
Autoimmune neurology is a rapidly expanding field driven by the discovery of neuroglial autoantibodies and encompassing a myriad of conditions affecting every level of the nervous system. Traditionally, autoantibodies targeting intracellular antigens are considered markers of T cell-mediated cytotoxicity, while those targeting extracellular antigens are viewed as pathogenic drivers of disease. However, recent advances highlight complex interactions between these immune mechanisms, suggesting a continuum of immunopathogenesis. The breakdown of immune tolerance, central to these conditions, is affected by modifiable and non-modifiable risk factors such as genetic predisposition, infections, and malignancy. While significant therapeutic advancements have revolutionized treatment of certain diseases, such as neuromyelitis optica, our understanding of many others, particularly T cell-mediated conditions, remains limited, with fewer treatment options available. Future research should focus on improving effector function modeling and deepening our understanding of the factors influencing immune tolerance, with the goal of providing novel treatment options and improving patient care.
Collapse
Affiliation(s)
- Yahel Segal
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - John Soltys
- Department of Neurosciences, Mayo Clinic, Jacksonville, FL, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Benjamin D S Clarkson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA
| | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA; Division of Experimental Neurology, Mayo Clinic, Rochester, MN, USA
| | - Sarosh R Irani
- Department of Neurosciences, Mayo Clinic, Jacksonville, FL, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA; Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals, Oxford, UK; Department of Neurology, Mayo Clinic, Jacksonville, FL, USA
| | - Sean J Pittock
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA; Center for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
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
|
4
|
Zhang L, Verkhratsky A, Shi FD. Astrocytes and microglia in multiple sclerosis and neuromyelitis optica. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:133-145. [PMID: 40148041 DOI: 10.1016/b978-0-443-19102-2.00001-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Multiple sclerosis and neuromyelitis optica are autoimmune neurodegenerative diseases primarily targeting myelin sheath and neuroglia. In multiple sclerosis, autoantibodies destroy oligodendrocytes and myelin, which underlies primary neurologic symptoms. Focal damage to myelin triggers reactive astrogliosis and microgliosis, which contribute to and to a large extent define the disease progression. In neuromyelitis optica, autoantibodies against water channel aquaporin 4 (AQP4), which are localized at astrocytic endfeet mediate damage of the glia limitans thus facilitating infiltration of blood-borne molecules and cells that propagate the damage to nerves and neurons. This primary astrocytopathy recruits microglia, which contribute to the neuroinflammatory response. Neuroglial cells therefore are potential targets for cell-specific therapies.
Collapse
Affiliation(s)
- Linjie Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Guo YC, Fu ZY, Ding ZJ. Immune infiltration associated C1q acts as a novel prognostic biomarker of cutaneous melanoma. Medicine (Baltimore) 2023; 102:e33088. [PMID: 36897727 PMCID: PMC9997796 DOI: 10.1097/md.0000000000033088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
C1q (complement C1q A chain, complement C1q B chain, and complement C1q C chain) is a recognized component of the classical complement pathway that influences the prognosis of various cancers. However, the effects of C1q on cutaneous melanoma (SKCM) outcomes and immune infiltration remain unknown. Gene expression profiling interactive analysis 2 and the human protein atlas were used to evaluate differential expression of C1q mRNA and protein. The relationship between C1q expression and clinicopathological features was also examined. The genetic alterations of C1q and their impact on survival were analyzed using the cbioportal database. The Kaplan-Meier approach was used to assess the significance of C1q in individuals with SKCM. The cluster profiler R package and the cancer single-cell state atlas database were used to investigate the function and mechanism of C1q in SKCM. The relationship between C1q and immune cell infiltration was estimated using single-sample gene set enrichment analysis. C1q expression was increased, and predicted a favorable prognosis. High C1q expression correlated with clinicopathological T stage, pathological stage, overall survival, and disease specific survival events. Moreover, C1q genetic alterations range from 2.7% to 4%, with no impact on prognosis. According to the enrichment analysis, C1q and immune-related pathways were closely connected. The link between complement C1q B chain and the functional state of inflammation was determined using the cancer single-cell state atlas database. In particular, C1q expression was significantly associated with infiltration of most immune cells and checkpoints PDCD1, CD274, and HAVCR2. The results of this study suggest that C1q is associated with prognosis and immune cell infiltration, supporting its value as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Yi-Cheng Guo
- Dermatology Hospital of Jiangxi Province, Nanchang, China
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Nanchang, Jiangxi, China
| | - Zhi-Yuan Fu
- Dermatology Hospital of Jiangxi Province, Nanchang, China
| | - Zhi-Jun Ding
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
| |
Collapse
|
6
|
Yick LW, Ma OKF, Chan EYY, Yau KX, Kwan JSC, Chan KH. T follicular helper cells contribute to pathophysiology in a model of neuromyelitis optica spectrum disorders. JCI Insight 2023; 8:161003. [PMID: 36649074 PMCID: PMC9977492 DOI: 10.1172/jci.insight.161003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) are inflammatory autoimmune disorders of the CNS. IgG autoantibodies targeting the aquaporin-4 water channel (AQP4-IgGs) are the pathogenic effector of NMOSD. Dysregulated T follicular helper (Tfh) cells have been implicated in loss of B cell tolerance in autoimmune diseases. The contribution of Tfh cells to disease activity and therapeutic potential of targeting these cells in NMOSD remain unclear. Here, we established an autoimmune model of NMOSD by immunizing mice against AQP4 via in vivo electroporation. After AQP4 immunization, mice displayed AQP4 autoantibodies in blood circulation, blood-brain barrier disruption, and IgG infiltration in spinal cord parenchyma. Moreover, AQP4 immunization induced motor impairments and NMOSD-like pathologies, including astrocytopathy, demyelination, axonal loss, and microglia activation. These were associated with increased splenic Tfh, Th1, and Th17 cells; memory B cells; and plasma cells. Aqp4-deficient mice did not display motor impairments and NMOSD-like pathologies after AQP4 immunization. Importantly, abrogating ICOS/ICOS-L signaling using anti-ICOS-L antibody depleted Tfh cells and suppressed the response of Th1 and Th17 cells, memory B cells, and plasma cells in AQP4-immunized mice. These findings were associated with ameliorated motor impairments and spinal cord pathologies. This study suggests a role of Tfh cells in the pathophysiology of NMOSD in a mouse model with AQP4 autoimmunity and provides an animal model for investigating the immunological mechanisms underlying AQP4 autoimmunity and developing therapeutic interventions targeting autoimmune reactions in NMOSD.
Collapse
|
7
|
Li H, Yang M, Song H, Sun M, Zhou H, Fu J, Zhou D, Bai W, Chen B, Lai M, Kang H, Wei S. ACT001 Relieves NMOSD Symptoms by Reducing Astrocyte Damage with an Autoimmune Antibody. Molecules 2023; 28:molecules28031412. [PMID: 36771078 PMCID: PMC9918908 DOI: 10.3390/molecules28031412] [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: 12/25/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a central nervous system inflammatory demyelinating disease, the pathogenesis of which involves autoantibodies targeting the extracellular epitopes of aquaporin-4 on astrocytes. We neutralized the AQP4-IgG from NMOSD patient sera using synthesized AQP4 extracellular epitope peptides and found that the severe cytotoxicity produced by aquaporin-4 immunoglobin (AQP4-IgG) could be blocked by AQP4 extracellular mimotope peptides of Loop A and Loop C in astrocyte protection and animal models. ACT001, a natural compound derivative, has shown anti-tumor activity in various cancers. In our study, the central nervous system anti-inflammatory effect of ACT001 was investigated. The results demonstrated the superior astrocyte protection activity of ACT001 at 10 µM. Furthermore, ACT001 decreases the behavioral score in the mouse NMOSD model, which was not inferior to Methylprednisolone Sodium Succinate, the first-line therapy of NMOSD in clinical practice. In summary, our study showed that astrocytes are protected by specific peptides, or small molecular drugs, which is a new strategy for the treatment of NMOSD. It is possible for ACT001 to be a promising therapy for NMOSD.
Collapse
Affiliation(s)
- Hongen Li
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Mo Yang
- Department of Neuro-Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Honglu Song
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
- Department of Ophthalmology, The 980th Hospital of the Chinese PLA Joint Logistics Support Force, Shijiazhuang 050082, China
| | - Mingming Sun
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Huanfen Zhou
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Junxia Fu
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Di Zhou
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Wenhao Bai
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Biyue Chen
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
| | - Mengying Lai
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
- Department of Public Health and Preventive Medicine, Shantou University Medical College, Shantou 515041, China
| | - Hao Kang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
- Correspondence: (H.K.); (S.W.)
| | - Shihui Wei
- Department of Ophthalmology, The Chinese People’s Liberation Army General Hospital & The Chinese People’s Liberation Army Medical School, Beijing 100853, China
- Correspondence: (H.K.); (S.W.)
| |
Collapse
|
8
|
Neuromyelitis Optica Spectrum Disorder: From Basic Research to Clinical Perspectives. Int J Mol Sci 2022; 23:ijms23147908. [PMID: 35887254 PMCID: PMC9323454 DOI: 10.3390/ijms23147908] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 02/05/2023] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory disease of the central nervous system characterized by relapses and autoimmunity caused by antibodies against the astrocyte water channel protein aquaporin-4. Over the past decade, there have been significant advances in the biologic knowledge of NMOSD, which resulted in the IDENTIFICATION of variable disease phenotypes, biomarkers, and complex inflammatory cascades involved in disease pathogenesis. Ongoing clinical trials are looking at new treatments targeting NMOSD relapses. This review aims to provide an update on recent studies regarding issues related to NMOSD, including the pathophysiology of the disease, the potential use of serum and cerebrospinal fluid cytokines as disease biomarkers, the clinical utilization of ocular coherence tomography, and the comparison of different animal models of NMOSD.
Collapse
|
9
|
Complement as a powerful "influencer" in the brain during development, adulthood and neurological disorders. Adv Immunol 2021; 152:157-222. [PMID: 34844709 DOI: 10.1016/bs.ai.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The complement system was long considered as only a powerful effector arm of the immune system that, while critically protective, could lead to inflammation and cell death if overactivated, even in the central nervous system (CNS). However, in the past decade it has been recognized as playing critical roles in key physiological processes in the CNS, including neurogenesis and synaptic remodeling in the developing and adult brain. Inherent in these processes are the interactions with cells in the brain, and the cascade of interactions and functional consequences that ensue. As a result, investigations of therapeutic approaches for both suppressing excessive complement driven neurotoxicity and aberrant sculpting of neuronal circuits, require broad (and deep) knowledge of the functional activities of multiple components of this highly evolved and regulated system to avoid unintended negative consequences in the clinic. Advances in basic science are beginning to provide a roadmap for translation to therapeutics, with both small molecule and biologics. Here, we present examples of the critical roles of proper complement function in the development and sculpting of the nervous system, and in enabling rapid protection from infection and clearance of dying cells. Microglia are highlighted as important command centers that integrate signals from the complement system and other innate sensors that are programed to provide support and protection, but that direct detrimental responses to aberrant activation and/or regulation of the system. Finally, we present promising research areas that may lead to effective and precision strategies for complement targeted interventions to promote neurological health.
Collapse
|
10
|
Cytoprotective IgG antibodies in sera from a subset of patients with AQP4-IgG seropositive neuromyelitis optica spectrum disorder. Sci Rep 2021; 11:21962. [PMID: 34753987 PMCID: PMC8578624 DOI: 10.1038/s41598-021-01294-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 10/26/2021] [Indexed: 11/09/2022] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system. Most NMOSD patients are seropositive for immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4), called AQP4-IgG. AQP4-IgG binding to aquaporin-4 causes complement-dependent cytotoxicity (CDC), leading to inflammation and demyelination. Here, CDC was measured in AQP4-expressing cells exposed to human complement and heat-inactivated sera from 108 AQP4-IgG seropositive NMOSD subjects and 25 non-NMOSD controls. AQP4-IgG positive sera produced a wide range of CDC, with 50% maximum cytotoxicity produced by as low as 0.2% serum concentration. Unexpectedly, 58 samples produced no cytotoxicity, and of those, four sera were cytoprotective against cytotoxic AQP4-IgG. Cytoprotection was found against different cytotoxic monoclonal AQP4-IgGs and NMOSD patient sera, and in primary astrocyte cultures. Mechanistic studies revealed that the protective factor is an IgG antibody that did not inhibit complement directly, but interfered with binding of cytotoxic AQP4-IgG to AQP4 and consequent C1q binding and complement activation. Further studies suggested that non-pathogenic AQP4-IgG, perhaps with altered glycosylation, may contribute to reduced or ineffectual binding of cytotoxic AQP4-IgG, as well as reduced cell-surface AQP4. The presence of natural cytoprotective antibodies in AQP4-IgG seropositive sera reveals an added level of complexity in NMOSD disease pathogenesis, and suggests the potential therapeutic utility of ‘convalescent’ serum or engineered protective antibody to interfere with pathogenic antibody in AQP4-IgG seropositive NMOSD.
Collapse
|
11
|
Li J, Bazzi SA, Schmitz F, Tanno H, McDaniel JR, Lee CH, Joshi C, Kim JE, Monson N, Greenberg BM, Hedfalk K, Melamed E, Ippolito GC. Molecular Level Characterization of Circulating Aquaporin-4 Antibodies in Neuromyelitis Optica Spectrum Disorder. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/5/e1034. [PMID: 34168058 PMCID: PMC8225010 DOI: 10.1212/nxi.0000000000001034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/27/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To determine whether distinct aquaporin-4 (AQP4)-IgG lineages play a role in neuromyelitis optica spectrum disorder (NMOSD) pathogenesis, we profiled the AQP4-IgG polyclonal serum repertoire and identified, quantified, and functionally characterized distinct AQP4-IgG lineages circulating in 2 patients with NMOSD. METHODS We combined high-throughput sequencing and quantitative immunoproteomics to simultaneously determine the constituents of both the B-cell receptor (BCR) and the serologic (IgG) anti-AQP4 antibody repertoires in the peripheral blood of patients with NMOSD. The monoclonal antibodies identified by this platform were recombinantly expressed and functionally characterized in vitro. RESULTS Multiple antibody lineages comprise serum AQP4-IgG repertoires. Their distribution, however, can be strikingly different in polarization (polyclonal vs pauciclonal). Among the 4 serum AQP4-IgG monoclonal antibodies we identified in 2 patients, 3 induced complement-dependent cytotoxicity in a model mammalian cell line (p < 0.01). CONCLUSIONS The composition and polarization of AQP4-IgG antibody repertoires may play an important role in NMOSD pathogenesis and clinical presentation. Here, we present a means of coupling both cellular (BCR) and serologic (IgG) antibody repertoire analysis, which has not previously been performed in NMOSD. Our analysis could be applied in the future to clinical management of patients with NMOSD to monitor disease activity over time as well as applied to other autoimmune diseases to facilitate a deeper understanding of disease pathogenesis relative to autoantibody clones.
Collapse
Affiliation(s)
- Jie Li
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Sam A Bazzi
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Florian Schmitz
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Hidetaka Tanno
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Jonathan R McDaniel
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Chang-Han Lee
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Chaitanya Joshi
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Jin Eyun Kim
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Nancy Monson
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Benjamin M Greenberg
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Kristina Hedfalk
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Esther Melamed
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX
| | - Gregory C Ippolito
- From the Department of Chemical Engineering (J.L., H.T., J.R.M., C.-H.L.), University of Texas at Austin, TX; Department of Neurology (S.A.B., E.M.), Dell Medical School, University of Texas at Austin, TX; Department of Chemistry & Molecular Biology (F.S., K.H.), University of Gothenburg, Sweden; Department of Neurology and Neurotherapeutics (C.J., N.M., B.M.G.), University of Texas Southwestern Medical Center, Dallas, TX; Department of Biomedical Engineering (J.E.K.), University of Texas at Austin, TX; and Department of Molecular Biosciences (G.C.I.), University of Texas at Austin, TX.
| |
Collapse
|
12
|
Tugizova M, Vlahovic L, Tomczak A, Wetzel NS, Han MH. New Therapeutic Landscape in Neuromyelitis Optica. Curr Treat Options Neurol 2021; 23:13. [PMID: 33814893 PMCID: PMC8008025 DOI: 10.1007/s11940-021-00667-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/11/2022]
Abstract
Purpose of review This review discusses the current treatment trends and emerging therapeutic landscape for patients with neuromyelitis optica spectrum disorder (NMOSD). Recent findings Conventional immune suppressive therapies, such as B cell depletion, have been used for long-term treatment. However, the availability of recent FDA-approved and investigational drugs has made therapeutic choices for NMOSD more complex. Summary Recent randomized clinical trials have shown that eculizumab, inebilizumab, and satralizumab are efficacious therapies for AQP4 seropositive NMOSD. These therapies may not have the same benefit in patients with seronegative NMOSD, including MOG-associated disease, and further investigation is required in this population. Reliable biomarkers to guide therapy decisions are urgently needed. There is a plethora of promising investigational therapies currently in the pipeline with exciting and novel mechanisms of action.
Collapse
Affiliation(s)
- Madina Tugizova
- Department of Neurology, Division of Neuroimmunology, Stanford University, 1201 Welch Road, MSLS p212, Stanford, CA 94305 USA.,Multiple Sclerosis Center, Stanford Hospital and Clinics, Palo Alto, CA USA
| | - Luka Vlahovic
- Department of Neurology, Creighton University School of Medicine, Omaha, NE USA
| | - Anna Tomczak
- Department of Neurology, Division of Neuroimmunology, Stanford University, 1201 Welch Road, MSLS p212, Stanford, CA 94305 USA.,Multiple Sclerosis Center, Stanford Hospital and Clinics, Palo Alto, CA USA
| | - Nora Sandrine Wetzel
- Department of Neurology, Division of Neuroimmunology, Stanford University, 1201 Welch Road, MSLS p212, Stanford, CA 94305 USA.,Faculty of Medicine, University of Zurich, Zürich, Switzerland
| | - May Htwe Han
- Department of Neurology, Division of Neuroimmunology, Stanford University, 1201 Welch Road, MSLS p212, Stanford, CA 94305 USA.,Multiple Sclerosis Center, Stanford Hospital and Clinics, Palo Alto, CA USA
| |
Collapse
|
13
|
Asavapanumas N, Tradtrantip L, Verkman AS. Targeting the complement system in neuromyelitis optica spectrum disorder. Expert Opin Biol Ther 2021; 21:1073-1086. [PMID: 33513036 DOI: 10.1080/14712598.2021.1884223] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Neuromyelitis optica spectrum disorder (NMOSD) is characterized by central nervous system inflammation and demyelination. In AQP4-IgG seropositive NMOSD, circulating immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4) cause tissue injury. Compelling evidence supports a pathogenic role for complement activation following AQP4-IgG binding to AQP4. Clinical studies supported the approval of eculizumab, an inhibitor of C5 cleavage, in AQP4-IgG seropositive NMOSD. AREAS COVERED This review covers in vitro, animal models, and human evidence for complement-dependent and complement-independent tissue injury in AQP4-IgG seropositive NMOSD. Complement targets are discussed, including complement proteins, regulators and anaphylatoxin receptors, and corresponding drug candidates. EXPERT OPINION Though preclinical data support a central pathogenic role of complement activation in AQP4-IgG seropositive NMOSD, they do not resolve the relative contributions of complement-dependent vs. complement-independent disease mechanisms such as antibody-dependent cellular cytotoxicity, T cell effector mechanisms, and direct AQP4-IgG-induced cellular injury. The best evidence that complement-dependent mechanisms predominate in AQP4-IgG seropositive NMOSD comes from eculizumab clinical data. Various drug candidates targeting distinct complement effector mechanisms may offer improved safety and efficacy. However, notwithstanding the demonstrated efficacy of complement inhibition in AQP4-IgG seropositive NMOSD, the ultimate niche for complement inhibition is not clear given multiple drug options with alternative mechanisms of action.Abbreviations: AAV2, Adeno-associated virus 2; ADCC, antibody-dependent cellular cytotoxicity; ANCA, antineutrophilic cytoplasmic autoantibody; AQP4, aquaporin-4; AQP4-IgG, AQP4-immunoglobulin G; C1-INH, C1-esterase inhibitor; C3aR, C3a receptor; C4BP, C4 binding protein; C5aR, C5a receptor; CDC, complement-dependent cytotoxicity; CFHR1, complement factor H related 1; CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; EndoS, endoglycosidase S; FHL-1, factor-H-like protein 1; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor protein-1; IgG, immunoglobulin G; IVIG, intravenous human immunoglobulin G; MAC, membrane attack complex; MBL, maltose-binding lectin; MBP, myelin basic protein; MOG, myelin oligodendrocyte glycoprotein; NK cell, natural killer cell; NMOSD, neuromyelitis optica spectrum disorder; OAP, orthogonal arrays of particles; PNH, paroxysmal nocturnal hemoglobinuria.
Collapse
Affiliation(s)
- Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
14
|
Chen T, Lennon VA, Liu YU, Bosco DB, Li Y, Yi MH, Zhu J, Wei S, Wu LJ. Astrocyte-microglia interaction drives evolving neuromyelitis optica lesion. J Clin Invest 2021; 130:4025-4038. [PMID: 32568214 DOI: 10.1172/jci134816] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
Neuromyelitis optica (NMO) is a severe inflammatory autoimmune CNS disorder triggered by binding of an IgG autoantibody to the aquaporin 4 (AQP4) water channel on astrocytes. Activation of cytolytic complement has been implicated as the major effector of tissue destruction that secondarily involves myelin. We investigated early precytolytic events in the evolving pathophysiology of NMO in mice by continuously infusing IgG (NMO patient serum-derived or AQP4-specific mouse monoclonal), without exogenous complement, into the spinal subarachnoid space. Motor impairment and sublytic NMO-compatible immunopathology were IgG dose dependent, AQP4 dependent, and, unexpectedly, microglia dependent. In vivo spinal cord imaging revealed a striking physical interaction between microglia and astrocytes that required signaling from astrocytes by the C3a fragment of their upregulated complement C3 protein. Astrocytes remained viable but lost AQP4. Previously unappreciated crosstalk between astrocytes and microglia involving early-activated CNS-intrinsic complement components and microglial C3a receptor signaling appears to be a critical driver of the precytolytic phase in the evolving NMO lesion, including initial motor impairment. Our results indicate that microglia merit consideration as a potential target for NMO therapeutic intervention.
Collapse
Affiliation(s)
| | - Vanda A Lennon
- Department of Neurology.,Department of Immunology, and.,Department of Laboratory Medicine/Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | | | | - Shihui Wei
- Department of Ophthalmology, Chinese PLA General Hospital, Beijing, China
| | - Long-Jun Wu
- Department of Neurology.,Department of Immunology, and.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
15
|
Abstract
Neuromyelitis optica (NMO) is a central nervous system (CNS) inflammatory autoimmune disease caused by antibodies against aquaporin-4 (AQP4) expressed on astrocytes. Binding of AQP4-specific antibodies (NMO-IgG) triggers activation of the complement cascade, which is responsible for astrocyte loss and secondary demyelination. Although the role for the cytolytic complement proteins in astrocyte destruction in NMO is well established, little is known regarding the initial phase of astrocyte injury. In this issue of the JCI, Chen and colleagues evaluated the precytolytic phase when NMO-IgG binds astrocytes in vivo in the absence of exogenous complement. NMO-IgG alone caused astrocyte activation and AQP4 loss. Surprisingly, microglia, CNS-resident innate immune cells that produce endogenous complement, were required for clinical manifestations of disease, a finding that suggests microglia may serve as a therapeutic target in NMO.
Collapse
|
16
|
Valencia-Sanchez C, Wingerchuk DM. Emerging Targeted Therapies for Neuromyelitis Optica Spectrum Disorders. BioDrugs 2020; 35:7-17. [PMID: 33301078 DOI: 10.1007/s40259-020-00460-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2020] [Indexed: 12/26/2022]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune, inflammatory disorder of the central nervous system that typically presents with recurrent episodes of optic neuritis, longitudinally extensive myelitis, brainstem, diencephalic, and cerebral syndromes. Up to 80% of NMOSD patients have a circulating pathogenic autoantibody that targets the water channel aquaporin-4 (AQP4-IgG). The discovery of AQP4-IgG transformed our understanding of the pathogenesis of the disease and its possible treatment targets. Monoclonal antibodies targeting terminal complement (eculizumab), CD19 (inebilizumab), and the interleukin-6 receptor (satralizumab) have demonstrated efficacy in NMOSD attack prevention in recent phase 3 trials and have gained subsequent regulatory approval in the USA and other countries. We aim to review the evidence supporting the efficacy of these new drugs.
Collapse
Affiliation(s)
| | - Dean M Wingerchuk
- Department of Neurology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ, 85259, USA.
| |
Collapse
|
17
|
Targeting the Initiator Protease of the Classical Pathway of Complement Using Fragment-Based Drug Discovery. Molecules 2020; 25:molecules25174016. [PMID: 32899120 PMCID: PMC7504721 DOI: 10.3390/molecules25174016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/23/2022] Open
Abstract
The initiating protease of the complement classical pathway, C1r, represents an upstream and pathway-specific intervention point for complement-related autoimmune and inflammatory diseases. Yet, C1r-targeted therapeutic development is currently underrepresented relative to other complement targets. In this study, we developed a fragment-based drug discovery approach using surface plasmon resonance (SPR) and molecular modeling to identify and characterize novel C1r-binding small-molecule fragments. SPR was used to screen a 2000-compound fragment library for binding to human C1r. This led to the identification of 24 compounds that bound C1r with equilibrium dissociation constants ranging between 160–1700 µM. Two fragments, termed CMP-1611 and CMP-1696, directly inhibited classical pathway-specific complement activation in a dose-dependent manner. CMP-1611 was selective for classical pathway inhibition, while CMP-1696 also blocked the lectin pathway but not the alternative pathway. Direct binding experiments mapped the CMP-1696 binding site to the serine protease domain of C1r and molecular docking and molecular dynamics studies, combined with C1r autoactivation assays, suggest that CMP-1696 binds within the C1r active site. The group of structurally distinct fragments identified here, along with the structure–activity relationship profiling of two lead fragments, form the basis for future development of novel high-affinity C1r-binding, classical pathway-specific, small-molecule complement inhibitors.
Collapse
|
18
|
Duchow A, Chien C, Paul F, Bellmann-Strobl J. Emerging drugs for the treatment of neuromyelitis optica. Expert Opin Emerg Drugs 2020; 25:285-297. [PMID: 32731771 DOI: 10.1080/14728214.2020.1803828] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Evidence-based treatment options for neuromyelitis optica spectrum disorders (NMOSD) patients are beginning to enter the market. Where previously, there was only the exclusive use of empiric and off-label immunosuppressants in this rare and devastating central nervous system autoimmune disease. AREAS COVERED In accordance to expanding pathogenetic insights, drugs in phase II and III clinical trials are presented in the context of the current treatment situation for acute attacks and immunopreventative strategies in NMOSD. Some such drugs are the 2019-approved complement inhibitor eculizumab, other compounds in late development include its modified successor ravulizumab, IL-6 receptor antibody satralizumab, CD19 targeting antibody inebilizumab and the TACI-Fc fusion protein telitacicept. EXPERT OPINION Moving from broad immunosuppression to tailored treatment strategies, the prospects for efficient NMOSD therapy are positive. For the first time in this disease, class I treatment evidence is available, but long-term data will be necessary to confirm the overall promising study results of the compounds close to approval. While drug development still centers around AQP4 antibody seropositive patients, current and future research requires consideration of possible diverging treatment demands for the smaller group of seronegative patients and patients with presence of MOG antibodies.
Collapse
Affiliation(s)
- Ankelien Duchow
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine , Berlin, Germany
| | - Claudia Chien
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany.,Department for Psychiatry and Psychotherapy - Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany
| | - Friedemann Paul
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine , Berlin, Germany
| | - Judith Bellmann-Strobl
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health , Berlin, Germany.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine , Berlin, Germany
| |
Collapse
|
19
|
Gong Y, Zhang YL, Wang Z, Song HH, Liu YC, Lv AW, Tian LL, Zhu WL, Fu Y, Ding XL, Cui LJ, Yan YP. Tanshinone IIA alleviates brain damage in a mouse model of neuromyelitis optica spectrum disorder by inducing neutrophil apoptosis. J Neuroinflammation 2020; 17:198. [PMID: 32586353 PMCID: PMC7318433 DOI: 10.1186/s12974-020-01874-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background Neuromyelitis optica spectrum disorder (NMOSD), an autoimmune astrocytopathic disease associated with the anti-aquaporin-4 (AQP4) antibody, is characterized by extensive necrotic lesions primarily located on the optic nerves and spinal cord. Tanshinone IIA (TSA), an active natural compound extracted from Salvia miltiorrhiza Bunge, has profound immunosuppressive effects on neutrophils. Objective The present study aimed to evaluate the effect of TSA on NMOSD mice and explore the underlying mechanisms. Mice were initially administered TSA (pre-TSA group, n = 20) or vehicle (vehicle group, n = 20) every 8 h for 3 days, and then NMOSD model was induced by intracerebral injection of NMOSD-immunoglobulin G (NMO-IgG) and human complement (hC). In addition, post-TSA mice (n = 10) were administered equal dose of TSA at 8 h and 16 h after model induction. At 24 h after intracerebral injection, histological analysis was performed to assess the inhibitory effects of TSA on astrocyte damage, demyelination, and neuroinflammation in NMOSD mice, and western blotting was conducted to clarify the effect of TSA on the NF-κB and MAPK signaling pathways. Furthermore, flow cytometry and western blotting were conducted to verify the proapoptotic effects of TSA on neutrophils in vitro. Results There was a profound reduction in astrocyte damage and demyelination in the pre-TSA group and post-TSA group. However, prophylactic administration of TSA induced a better effect than therapeutic treatment. The number of infiltrated neutrophils was also decreased in the lesions of NMOSD mice that were pretreated with TSA. We confirmed that prophylactic administration of TSA significantly promoted neutrophil apoptosis in NMOSD lesions in vivo, and this proapoptotic effect was mediated by modulating the caspase pathway in the presence of inflammatory stimuli in vitro. In addition, TSA restricted activation of the NF-κB signaling pathway in vivo. Conclusion Our data provide evidence that TSA can act as a prophylactic agent that reduces NMO-IgG-induced damage in the mouse brain by enhancing the resolution of inflammation by inducing neutrophil apoptosis, and TSA may serve as a promising therapeutic agent for neutrophil-associated inflammatory disorders, such as NMOSD.
Collapse
Affiliation(s)
- Ye Gong
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Ya-Ling Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Zhen Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.,Department of Neurology, Xuanwu Hospital, Capital Medical University, No.45, Changchun Street, Beijing, 100053, China
| | - Huan-Huan Song
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Yuan-Chu Liu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Ao-Wei Lv
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No.154, Anshan Road, Tianjin, 300052, China
| | - Li-Li Tian
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No.154, Anshan Road, Tianjin, 300052, China
| | - Wen-Li Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, No.154, Anshan Road, Tianjin, 300052, China
| | - Ying Fu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Xiao-Li Ding
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China
| | - Lang-Jun Cui
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.
| | - Ya-Ping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Xi'an, 710119, China.
| |
Collapse
|
20
|
Monoclonal Antibody-Based Treatments for Neuromyelitis Optica Spectrum Disorders: From Bench to Bedside. Neurosci Bull 2020; 36:1213-1224. [PMID: 32533450 DOI: 10.1007/s12264-020-00525-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/10/2020] [Indexed: 12/20/2022] Open
Abstract
Neuromyelitis optica (NMO)/NMO spectrum disorder (NMOSD) is a chronic, recurrent, antibody-mediated, inflammatory demyelinating disease of the central nervous system, characterized by optic neuritis and transverse myelitis. The binding of NMO-IgG with astrocytic aquaporin-4 (AQP4) functions directly in the pathogenesis of >60% of NMOSD patients, and causes astrocyte loss, secondary inflammatory infiltration, demyelination, and neuron death, potentially leading to paralysis and blindness. Current treatment options, including immunosuppressive agents, plasma exchange, and B-cell depletion, are based on small retrospective case series and open-label studies. It is noteworthy that monoclonal antibody (mAb) therapy is a better option for autoimmune diseases due to its high efficacy and tolerability. Although the pathophysiological mechanisms of NMOSD remain unknown, increasingly, therapeutic studies have focused on mAbs, which target B cell depletion, complement and inflammation cascade inactivation, blood-brain-barrier protection, and blockade of NMO-IgG-AQP4 binding. Here, we review the targets, characteristics, mechanisms of action, development, and potential efficacy of mAb trials in NMOSD, including preclinical and experimental investigations.
Collapse
|
21
|
Duchow A, Paul F, Bellmann-Strobl J. Current and emerging biologics for the treatment of neuromyelitis optica spectrum disorders. Expert Opin Biol Ther 2020; 20:1061-1072. [DOI: 10.1080/14712598.2020.1749259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Ankelien Duchow
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Friedemann Paul
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Judith Bellmann-Strobl
- Neurocure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
22
|
Tradtrantip L, Asavapanumas N, Verkman AS. Emerging therapeutic targets for neuromyelitis optica spectrum disorder. Expert Opin Ther Targets 2020; 24:219-229. [PMID: 32070155 DOI: 10.1080/14728222.2020.1732927] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelinating disease of the central nervous system affecting primarily the spinal cord and optic nerves. Most NMOSD patients are seropositive for immunoglobulin G autoantibodies against astrocyte water channel aquaporin-4, called AQP4-IgG, which cause astrocyte injury leading to demyelination and neurological impairment. Current therapy for AQP4-IgG seropositive NMOSD includes immunosuppression, B cell depletion, and plasma exchange. Newer therapies target complement, CD19 and IL-6 receptors.Areas covered: This review covers early-stage pre-clinical therapeutic approaches for seropositive NMOSD. Targets include pathogenic AQP4-IgG autoantibodies and their binding to AQP4, complement-dependent and cell-mediated cytotoxicity, blood-brain barrier, remyelination and immune effector and regulatory cells, with treatment modalities including small molecules, biologics, and cells.Expert opinion: Though newer NMOSD therapies appear to have increased efficacy in reducing relapse rate and neurological deficit, increasingly targeted therapies could benefit NMOSD patients with ongoing relapses and could potentially be superior in efficacy and safety. Of the various early-stage therapeutic approaches, IgG inactivating enzymes, aquaporumab blocking antibodies, drugs targeting early components of the classical complement system, complement regulator-targeted drugs, and Fc-based multimers are of interest. Curative strategies, perhaps involving AQP4 tolerization, remain intriguing future possibilities.
Collapse
Affiliation(s)
- Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
23
|
Duan T, Tradtrantip L, Phuan PW, Bennett JL, Verkman AS. Affinity-matured 'aquaporumab' anti-aquaporin-4 antibody for therapy of seropositive neuromyelitis optica spectrum disorders. Neuropharmacology 2019; 162:107827. [PMID: 31654702 DOI: 10.1016/j.neuropharm.2019.107827] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 10/25/2022]
Abstract
Pathogenesis in seropositive neuromyelitis optica spectrum disorders (herein called NMO) involves binding of IgG1 autoantibodies to aquaporin-4 (AQP4) on astrocytes in the central nervous system, which initiates complement and cellular injury. We previously developed an antibody blocking approach for potential therapy of NMO in which an engineered, monoclonal, anti-AQP4 antibody lacking cytotoxicity effector functions (called aquaporumab) blocked binding of NMO autoantibodies to astrocyte AQP4 (Tradtrantip et al. Ann. Neurol. 71, 314-322, 2012). Here, a high-affinity aquaporumab, which was generated by affinity maturation using saturation mutagenesis, was shown to block cellular injury caused by NMO patient sera. Anti-AQP4 antibody rAb-53, a fully human antibody with effector function neutralizing Fc mutations L234A/L235A and affinity-enhancing Fab mutations Y50R/S56R, called AQmabAM, bound to AQP4 in cell cultures with Kd ~ 18 ng/ml (~0.12 nM), ~8-fold greater affinity than the original antibody. AQmabAM, but without L234A/L235A Fc mutations, produced complement-dependent cytotoxicity (CDC) with EC50 ~ 82 ng/ml. AQmabAM prevented CDC produced by sera from eight NMO patients with IC50 ranging from 40 to 80 ng/ml, and similarly prevented antibody-dependent cellular cytotoxicity (ADCC). Mechanistic studies demonstrated that AQmabAM blocked binding of serum NMO autoantibodies to AQP4. AQmabAM offers a targeted, non-immunosuppressive approach for therapy of seropositive NMO. Autoantibody blocking may be a useful therapeutic strategy for other autoimmune diseases as well.
Collapse
Affiliation(s)
- Tianjiao Duan
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143, USA; Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143, USA
| | - Puay-Wah Phuan
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143, USA
| | - Jeffrey L Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado Anschutz Medical Campus, Denver, CO, 80045, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
24
|
Duan T, Verkman AS. Experimental animal models of aquaporin-4-IgG-seropositive neuromyelitis optica spectrum disorders: progress and shortcomings. Brain Pathol 2019; 30:13-25. [PMID: 31587392 DOI: 10.1111/bpa.12793] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022] Open
Abstract
Neuromyelitis optica spectrum disorders (NMOSD) is a heterogeneous group of neuroinflammatory conditions associated with demyelination primarily in spinal cord and optic nerve, and to a lesser extent in brain. Most NMOSD patients are seropositive for IgG autoantibodies against aquaporin-4 (AQP4-IgG), the principal water channel in astrocytes. There has been interest in establishing experimental animal models of seropositive NMOSD (herein referred to as NMO) in order to elucidate NMO pathogenesis mechanisms and to evaluate drug candidates. An important outcome of early NMO animal models was evidence for a pathogenic role of AQP4-IgG. However, available animal models of NMO, based largely on passive transfer to rodents of AQP4-IgG or transfer of AQP4-sensitized T cells, often together with pro-inflammatory maneuvers, only partially recapitulate the clinical and pathological features of human NMO, and are inherently biased toward humoral or cellular immune mechanisms. This review summarizes current progress and shortcomings in experimental animal models of seropositive NMOSD, and opines on the import of advancing animal models.
Collapse
Affiliation(s)
- Tianjiao Duan
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143.,Department of Neurology, Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, 94143
| |
Collapse
|
25
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
26
|
Zhu W, Wang Z, Hu S, Gong Y, Liu Y, Song H, Ding X, Fu Y, Yan Y. Human C5-specific single-chain variable fragment ameliorates brain injury in a model of NMOSD. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e561. [PMID: 31044149 PMCID: PMC6467685 DOI: 10.1212/nxi.0000000000000561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/05/2019] [Indexed: 12/01/2022]
Abstract
Objective Using phage display, we sought to screen single-chain variable fragments (scFvs) against complement C5 to treat neuromyelitis optica spectrum disorder (NMOSD). Methods After 5 rounds of phage display, we isolated individual clones and identified phage clones specifically binding to C5 using ELISA. Using aquaporin-4 (AQP4)-transfected cells in vitro, we confirmed whether these scFvs prevented complement-dependent cytotoxicity (CDC) caused by the serum of patients with NMOSD and human complement (hC). We selected an NMOSD mouse model, in which intracerebral NMOSD immunoglobulin G (IgG) and hC injections induce NMOSD-like lesions in vivo. Results We obtained scFvs to test specificity and blocking efficiency. The scFv C5B3 neutralized C5 in the complement activation pathway, which prevented AQP4-IgG-mediated CDC in AQP4-transfected cells. In an NMOSD mouse model, C5B3 prevented AQP4 and astrocyte loss, decreased demyelination, and reduced inflammatory infiltration and membrane attack complex formation in lesions. Conclusions We used phage display to screen C5B3 against C5, which was effective in inhibiting cytotoxicity in vitro and preventing CNS pathology in vivo.
Collapse
Affiliation(s)
- Wenli Zhu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhen Wang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Suying Hu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ye Gong
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuanchu Liu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Huanhuan Song
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Ding
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ying Fu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yaping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry (W.Z., Z.W., S.H., Y.G., Y.L., H.S., X.D., Y.F., Y.Y.), National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an; Department of Neurology (W.Z.), Tianjin Neurological Institute, Tianjin Medical University General Hospital; and Department of Neurology (Z.W.), Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
27
|
Soltys J, Liu Y, Ritchie A, Wemlinger S, Schaller K, Schumann H, Owens GP, Bennett JL. Membrane assembly of aquaporin-4 autoantibodies regulates classical complement activation in neuromyelitis optica. J Clin Invest 2019; 129:2000-2013. [PMID: 30958797 DOI: 10.1172/jci122942] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 02/26/2019] [Indexed: 01/29/2023] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune CNS disorder mediated by pathogenic aquaporin-4 (AQP4) water channel autoantibodies (AQP4-IgG). Although AQP4-IgG-driven complement-dependent cytotoxicity (CDC) is critical for the formation of NMO lesions, the molecular mechanisms governing optimal classical pathway activation are unknown. We investigated the molecular determinants driving CDC in NMO using recombinant AQP4-specific autoantibodies (AQP4 rAbs) derived from affected patients. We identified a group of AQP4 rAbs targeting a distinct extracellular loop C epitope that demonstrated enhanced CDC on target cells. Targeted mutations of AQP4 rAb Fc domains that enhance or diminish C1q binding or antibody Fc-Fc interactions showed that optimal CDC was driven by the assembly of multimeric rAb platforms that increase multivalent C1q binding and facilitate C1q activation. A peptide that blocks antibody Fc-Fc interaction inhibited CDC induced by AQP4 rAbs and polyclonal NMO patient sera. Super-resolution microscopy revealed that AQP4 rAbs with enhanced CDC preferentially formed organized clusters on supramolecular AQP4 orthogonal arrays, linking epitope-dependent multimeric assembly with enhanced C1q binding and activation. The resulting model of AQP4-IgG CDC provides a framework for understanding classical complement activation in human autoantibody-mediated disorders and identifies a potential new therapeutic avenue for treating NMO.
Collapse
Affiliation(s)
- John Soltys
- Neuroscience and Medical Scientist Training Programs
| | | | | | | | | | | | | | - Jeffrey L Bennett
- Neuroscience and Medical Scientist Training Programs.,Department of Neurology, and.,Department of Ophthalmology, University of Colorado at Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
28
|
CD55 upregulation in astrocytes by statins as potential therapy for AQP4-IgG seropositive neuromyelitis optica. J Neuroinflammation 2019; 16:57. [PMID: 30851734 PMCID: PMC6408857 DOI: 10.1186/s12974-019-1448-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Background Neuromyelitis optica spectrum disorder (herein called NMO) is an inflammatory demyelinating disease that can be initiated by binding of immunoglobulin G autoantibodies (AQP4-IgG) to aquaporin-4 on astrocytes, causing complement-dependent cytotoxicity (CDC) and downstream inflammation. The increased NMO pathology in rodents deficient in complement regulator protein CD59 following passive transfer of AQP4-IgG has suggested the potential therapeutic utility of increasing the expression of complement regulator proteins. Methods A cell-based ELISA was developed to screen for pharmacological upregulators of endogenous CD55 and CD59 in a human astrocyte cell line. A statin identified from the screen was characterized in cell culture models and rodents for its action on complement regulator protein expression and its efficacy in models of seropositive NMO. Results Screening of ~ 11,500 approved and investigational drugs and nutraceuticals identified transcriptional upregulators of CD55 but not of CD59. Several statins, including atorvastatin, simvastatin, lovastatin, and fluvastatin, increased CD55 protein expression in astrocytes, including primary cultures, by three- to four-fold at 24 h, conferring significant protection against AQP4-IgG-induced CDC. Mechanistic studies revealed that CD55 upregulation involves inhibition of the geranylgeranyl transferase pathway rather than inhibition of cholesterol biosynthesis. Oral atorvastatin at 10–20 mg/kg/day for 3 days strongly increased CD55 immunofluorescence in mouse brain and spinal cord and reduced NMO pathology following intracerebral AQP4-IgG injection. Conclusion Atorvastatin or other statins may thus have therapeutic benefit in AQP4-IgG seropositive NMO by increasing CD55 expression, in addition to their previously described anti-inflammatory and immunomodulatory actions.
Collapse
|
29
|
Bheemareddy BR, Pulipeta M, Iyer P, Dirisala VR. Effect of the total galactose content on complement-dependent cytotoxicity of the therapeutic anti-CD20 IgG1 antibodies under temperature stress conditions. J Carbohydr Chem 2019. [DOI: 10.1080/07328303.2018.1541995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
| | | | - Pradeep Iyer
- R&D Division, Hetero Biopharma Limited, Mahaboob Nagar, Telangana, India
| | - Vijaya R. Dirisala
- Department of Biotechnology, Vignan’s Foundation for Science, Technology and Research (VFSTR), Guntur, Andhra Pradesh, India
| |
Collapse
|
30
|
Collongues N, Ayme-Dietrich E, Monassier L, de Seze J. Pharmacotherapy for Neuromyelitis Optica Spectrum Disorders: Current Management and Future Options. Drugs 2019; 79:125-142. [PMID: 30623348 DOI: 10.1007/s40265-018-1039-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuromyelitis optica (NMO) is an inflammatory and demyelinating disease of the central nervous system. Although the prevalence of NMO is low, the rapid and severe impairment observed in patients has led to extensive development of research in the fields of diagnostic criteria and therapy in the past 15 years. With improved understanding of the pathophysiology of NMO and the role of aquaporin-4 (AQP4) or myelin oligodendrocyte glycoprotein antibodies, numerous therapeutic approaches have been proposed and are currently undergoing evaluation. In this review, we describe the rationale for existing therapeutics and their benefit/risk ratio. We also discuss the pharmacological and clinical interest of future approaches targeting, among others, B or T cells, the blood-central nervous system barrier, complement, polynuclear cells, AQP4-antibody linkage and AQP4 activity. The numerous agents under development are the result of a major collaborative effort all over the world. After the considerable progress on diagnosis, we are now close to class I evidence for a therapeutic effect of several drugs in NMO spectrum disorders, most notably with the anti-interleukin-6 receptor antibody (satralizumab) and anti-complement-5 antibody (eculizumab).
Collapse
Affiliation(s)
- Nicolas Collongues
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67000, Strasbourg, France.
- Département de Neurologie, Centre Hospitalier Universitaire de Strasbourg, Avenue Molière, 67200, Strasbourg, France.
- Centre d'investigation Clinique, INSERM U1434, Centre Hospitalier Universitaire de Strasbourg, 1 Place de l'Hôpital, 67000, Strasbourg, France.
| | - Estelle Ayme-Dietrich
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, Fédération de Médecine Translationnelle, Faculté de Médecine, Université de Strasbourg, 11 rue Humann, 67000, Strasbourg, France
| | - Laurent Monassier
- Laboratoire de Pharmacologie et Toxicologie Neurocardiovasculaire, Fédération de Médecine Translationnelle, Faculté de Médecine, Université de Strasbourg, 11 rue Humann, 67000, Strasbourg, France
| | - Jérôme de Seze
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67000, Strasbourg, France
- Département de Neurologie, Centre Hospitalier Universitaire de Strasbourg, Avenue Molière, 67200, Strasbourg, France
- Centre d'investigation Clinique, INSERM U1434, Centre Hospitalier Universitaire de Strasbourg, 1 Place de l'Hôpital, 67000, Strasbourg, France
| |
Collapse
|
31
|
Dobó J, Kocsis A, Gál P. Be on Target: Strategies of Targeting Alternative and Lectin Pathway Components in Complement-Mediated Diseases. Front Immunol 2018; 9:1851. [PMID: 30135690 PMCID: PMC6092519 DOI: 10.3389/fimmu.2018.01851] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022] Open
Abstract
The complement system has moved into the focus of drug development efforts in the last decade, since its inappropriate or uncontrolled activation has been recognized in many diseases. Some of them are primarily complement-mediated rare diseases, such as paroxysmal nocturnal hemoglobinuria, C3 glomerulonephritis, and atypical hemolytic uremic syndrome. Complement also plays a role in various multifactorial diseases that affect millions of people worldwide, such as ischemia reperfusion injury (myocardial infarction, stroke), age-related macular degeneration, and several neurodegenerative disorders. In this review, we summarize the potential advantages of targeting various complement proteins with special emphasis on the components of the lectin (LP) and the alternative pathways (AP). The serine proteases (MASP-1/2/3, factor D, factor B), which are responsible for the activation of the cascade, are straightforward targets of inhibition, but the pattern recognition molecules (mannose-binding lectin, other collectins, and ficolins), the regulatory components (factor H, factor I, properdin), and C3 are also subjects of drug development. Recent discoveries about cross-talks between the LP and AP offer new approaches for clinical intervention. Mannan-binding lectin-associated serine proteases (MASPs) are not just responsible for LP activation, but they are also indispensable for efficient AP activation. Activated MASP-3 has recently been shown to be the enzyme that continuously supplies factor D (FD) for the AP by cleaving pro-factor D (pro-FD). In this aspect, MASP-3 emerges as a novel feasible target for the regulation of AP activity. MASP-1 was shown to be required for AP activity on various surfaces, first of all on LPS of Gram-negative bacteria.
Collapse
Affiliation(s)
- József Dobó
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andrea Kocsis
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
32
|
Fang L, Wang Z, Song HH, Zhou YF, Chen C, Yan YP, Qiu W. Hydroxychloroquine fails to attenuate lesion development in a mouse model of neuromyelitis optica. CNS Neurosci Ther 2018; 24:842-845. [PMID: 29761646 DOI: 10.1111/cns.12975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Affiliation(s)
- Ling Fang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhen Wang
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Huan-Huan Song
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yi-Fan Zhou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ya-Ping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
33
|
Motamed-Gorji N, Matin N, Tabatabaie O, Pavone P, Romano C, Falsaperla R, Vitaliti G. Biological Drugs in Guillain-Barré Syndrome: An Update. Curr Neuropharmacol 2018; 15:938-950. [PMID: 27964705 PMCID: PMC5652014 DOI: 10.2174/1570159x14666161213114904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background: Guillain-Barré Syndrome (GBS) is currently considered the most common global cause of acute flaccid paralysis. Currently, standard therapy for Guillain-Barré Syndrome includes intravenous immunoglobulin or plasma exchange. Despite medical advances regarding these treatments, many treated patients do not reach full recovery. Therefore several biological agents have attracted the attentions from researchers during the last decades, and various studies have investigated their role in Guillain-Barré Syndrome. Objective: The present study aims to address emerging biological approaches to GBS while considering their efficiency and safety in treating the disease. Materials and Methods: An extensive electronic literature search was conducted by two researchers from April 2016 to July 2016. Original articles, clinical trials, systematic reviews (with or without meta-analysis) and case reports were selected. Titles and abstracts of papers were screened by reviewers to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. Results: Herein authors focused on the literature data concerning emerging biological therapeutic agents, namely anti-C5 monoclonal antibody (Eculizumab), anti-C1q monoclonal antibody, anti-T cell monoclonal antibody, anti-CD2 monoclonal antibody, anti L-selectin monoclonal antibody, anti-CD20 monoclonal antibody (Rituximab), anti-CD52 monoclonal antibody (Alemtuzumab) and cytokine targets. By far, none of these agents have been approved for the treatment of GBS by FDA. Conclusion: Literature findings represented in current review herald promising results for using these biological targets. Current review represents a summary of what is already in regards and what progress is required to improve the immunotherapeutic approach of treating GBS via future studies.
Collapse
Affiliation(s)
| | - Nassim Matin
- Department of Neurology, Massachusetts General Hospital, Boston, MA. United States
| | - Omidreza Tabatabaie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. United States
| | - Piero Pavone
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Catia Romano
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Raffaele Falsaperla
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Giovanna Vitaliti
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| |
Collapse
|
34
|
Tradtrantip L, Felix CM, Spirig R, Morelli AB, Verkman A. Recombinant IgG1 Fc hexamers block cytotoxicity and pathological changes in experimental in vitro and rat models of neuromyelitis optica. Neuropharmacology 2018; 133:345-353. [PMID: 29428821 PMCID: PMC6322534 DOI: 10.1016/j.neuropharm.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/17/2022]
Abstract
Intravenous human immunoglobulin G (IVIG) may have therapeutic benefit in neuromyelitis optica spectrum disorders (herein called NMO), in part because of the anti-inflammatory properties of the IgG Fc region. Here, we evaluated recombinant Fc hexamers consisting of the IgM μ-tailpiece fused with the Fc region of human IgG1. In vitro, the Fc hexamers prevented cytotoxicity in aquaporin-4 (AQP4) expressing cells and in rat spinal cord slice cultures exposed to NMO anti-AQP4 autoantibody (AQP4-IgG) and complement, with >500-fold greater potency than IVIG or monomeric Fc fragments. Fc hexamers at low concentration also prevented antibody-dependent cellular cytotoxicity produced by AQP4-IgG and natural killer cells. Serum from rats administered a single intravenous dose of Fc hexamers at 50 mg/kg taken at 8 h did not produce complement-dependent cytotoxicity when added to AQP4-IgG-treated AQP4-expressing cell cultures. In an experimental rat model of NMO produced by intracerebral injection of AQP4-IgG, Fc hexamers at 50 mg/kg administered before and at 12 h after AQP4-IgG fully prevented astrocyte injury, complement activation, inflammation and demyelination. These results support the potential therapeutic utility of recombinant IgG1 Fc hexamers in AQP4-IgG seropositive NMO.
Collapse
Affiliation(s)
- Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | - Christian M. Felix
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | | | | | - A.S. Verkman
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| |
Collapse
|
35
|
Li Z, Han J, Ren H, Ma CG, Shi FD, Liu Q, Li M. Astrocytic Interleukin-15 Reduces Pathology of Neuromyelitis Optica in Mice. Front Immunol 2018; 9:523. [PMID: 29616032 PMCID: PMC5867910 DOI: 10.3389/fimmu.2018.00523] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/28/2018] [Indexed: 12/05/2022] Open
Abstract
Astrocyte loss induced by neuromyelitis optica (NMO)-IgG and complement-dependent cytotoxicity (CDC) is the hallmark of NMO pathology. The survival of astrocytes is thought to reflect astrocyte exposure to environmental factors in the CNS and the response of astrocytes to these factors. However, still unclear are how astrocytes respond to NMO-IgG and CDC, and what CNS environmental factors may impact the survival of astrocytes. In a murine model of NMO induced by intracerebral injection of NMO-IgG and human complement, we found dramatic upregulation of IL-15 in astrocytes. To study the role of astrocytic IL-15 in NMO, we generated a transgenic mouse line with targeted expression of IL-15 in astrocytes (IL-15tg), in which the expression of IL-15 is controlled by a glial fibrillary acidic protein promoter. We showed that astrocyte-targeted expression of IL-15 attenuates astrocyte injury and the loss of aquaporin-4 in the brain. Reduced blood–brain barrier leakage and immune cell infiltration are also found in the lesion of IL-15tg mice subjected to NMO induction. IL-15tg astrocytes are less susceptible to NMO-IgG-mediated CDC than their wild-type counterparts. The enhanced resistance of IL-15tg astrocytes to cytotoxicity and cell death involves NF-κB signaling pathway. Our findings suggest that IL-15 reduces astrocyte loss and NMO pathology.
Collapse
Affiliation(s)
- Zhiguo Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinrui Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cun-Gen Ma
- Shanxi University of Traditional Chinese Medicine, Taiyuan, China
| | - Fu-Dong Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
36
|
|
37
|
Soltys JN, Meyer SA, Schumann H, Gibson EA, Restrepo D, Bennett JL. Determining the Spatial Relationship of Membrane-Bound Aquaporin-4 Autoantibodies by STED Nanoscopy. Biophys J 2017; 112:1692-1702. [PMID: 28445760 DOI: 10.1016/j.bpj.2017.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023] Open
Abstract
Determining the spatial relationship of individual proteins in dense assemblies remains a challenge for superresolution nanoscopy. The organization of aquaporin-4 (AQP4) into large plasma membrane assemblies provides an opportunity to image membrane-bound AQP4 antibodies (AQP4-IgG) and evaluate changes in their spatial distribution due to alterations in AQP4 isoform expression and AQP4-IgG epitope specificity. Using stimulated emission depletion nanoscopy, we imaged secondary antibody labeling of monoclonal AQP4-IgGs with differing epitope specificity bound to isolated tetramers (M1-AQP4) and large orthogonal arrays of AQP4 (M23-AQP4). Imaging secondary antibodies bound to M1-AQP4 allowed us to infer the size of individual AQP4-IgG binding events. This information was used to model the assembly of larger AQP4-IgG complexes on M23-AQP4 arrays. A scoring algorithm was generated from these models to characterize the spatial arrangement of bound AQP4-IgG antibodies, yielding multiple epitope-specific patterns of bound antibodies on M23-AQP4 arrays. Our results delineate an approach to infer spatial relationships within protein arrays using stimulated emission depletion nanoscopy, offering insight into how information on single antibody fluorescence events can be used to extract information from dense protein assemblies under a biologic context.
Collapse
Affiliation(s)
- John N Soltys
- Medical Scientist Training and Neuroscience Graduate Training Programs, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Stephanie A Meyer
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hannah Schumann
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Emily A Gibson
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Diego Restrepo
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jeffrey L Bennett
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
38
|
Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends Mol Med 2017; 23:512-533. [DOI: 10.1016/j.molmed.2017.03.008] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/15/2017] [Accepted: 03/26/2017] [Indexed: 01/17/2023]
|
39
|
Hubbard JA, Szu JI, Binder DK. The role of aquaporin-4 in synaptic plasticity, memory and disease. Brain Res Bull 2017; 136:118-129. [PMID: 28274814 DOI: 10.1016/j.brainresbull.2017.02.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/25/2022]
Abstract
Since the discovery of aquaporins, it has become clear that the various mammalian aquaporins play critical physiological roles in water and ion balance in multiple tissues. Aquaporin-4 (AQP4), the principal aquaporin expressed in the central nervous system (CNS, brain and spinal cord), has been shown to mediate CNS water homeostasis. In this review, we summarize new and exciting studies indicating that AQP4 also plays critical and unanticipated roles in synaptic plasticity and memory formation. Next, we consider the role of AQP4 in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), neuromyelitis optica (NMO), epilepsy, traumatic brain injury (TBI), and stroke. Each of these conditions involves changes in AQP4 expression and/or distribution that may be functionally relevant to disease physiology. Insofar as AQP4 is exclusively expressed on astrocytes, these data provide new evidence of "astrocytopathy" in the etiology of diverse neurological diseases.
Collapse
Affiliation(s)
- Jacqueline A Hubbard
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Jenny I Szu
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States.
| |
Collapse
|
40
|
Bading H. Therapeutic targeting of the pathological triad of extrasynaptic NMDA receptor signaling in neurodegenerations. J Exp Med 2017; 214:569-578. [PMID: 28209726 PMCID: PMC5339681 DOI: 10.1084/jem.20161673] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/02/2017] [Accepted: 01/05/2017] [Indexed: 01/12/2023] Open
Abstract
Activation of extrasynaptic N-methyl-d-aspartate (NMDA) receptors causes neurodegeneration and cell death. The disease mechanism involves a pathological triad consisting of mitochondrial dysfunction, loss of integrity of neuronal structures and connectivity, and disruption of excitation-transcription coupling caused by CREB (cyclic adenosine monophosphate-responsive element-binding protein) shut-off and nuclear accumulation of class IIa histone deacetylases. Interdependency within the triad fuels an accelerating disease progression that culminates in failure of mitochondrial energy production and cell loss. Both acute and slowly progressive neurodegenerative conditions, including stroke, Alzheimer's disease, amyotrophic lateral sclerosis, and Huntington's disease, share increased death signaling by extrasynaptic NMDA receptors caused by elevated extracellular glutamate concentrations or relocalization of NMDA receptors to extrasynaptic sites. Six areas of therapeutic objectives are defined, based on which a broadly applicable combination therapy is proposed to combat the pathological triad of extrasynaptic NMDA receptor signaling that is common to many neurodegenerative diseases.
Collapse
Affiliation(s)
- Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Liu Y, Harlow DE, Given KS, Owens GP, Macklin WB, Bennett JL. Variable sensitivity to complement-dependent cytotoxicity in murine models of neuromyelitis optica. J Neuroinflammation 2016; 13:301. [PMID: 27905992 PMCID: PMC5134246 DOI: 10.1186/s12974-016-0767-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/21/2016] [Indexed: 01/06/2023] Open
Abstract
Background Studies of neuromyelitis optica (NMO), an autoimmune disease of the central nervous system (CNS), have demonstrated that autoantibodies against the water channel aquaporin-4 (AQP4) induce astrocyte damage through complement-dependent cytotoxicity (CDC). In developing experimental models of NMO using cells, tissues or animals from mice, co-administration of AQP4-IgG and normal human serum, which serves as the source of human complement (HC), is required. The sensitivity of mouse CNS cells to HC and CDC in these models is not known. Methods We used HC and recombinant monoclonal antibodies (rAbs) against AQP4 to investigate CDC on mouse neurons, astrocytes, differentiated oligodendrocytes (OLs), and oligodendrocyte progenitors (OPCs) in the context of purified monocultures, neuroglial mixed cultures, and organotypic cerebellar slices. Results We found that murine neurons, OLs, and OPCs were sensitive to HC in monocultures. In mixed murine neuroglial cultures, HC-mediated toxicity to neurons and OLs was reduced; however, astrocyte damage induced by an AQP-specific rAb #53 and HC increased neuronal and oligodendroglial loss. OPCs were resistant to HC toxicity in neuroglial mixed cultures. In mouse cerebellar slices, damage to neurons and OLs following rAb #53-mediated CDC was further reduced, but in contrast to neuroglial mixed cultures, astrocyte damage sensitized OPCs to complement damage. Finally, we established that some injury to neurons, OLs, and OPCs in cell and slice cultures resulted from the activation of HC by anti-tissue antibodies to mouse cells. Conclusions Murine neurons and oligodendroglia demonstrate variable sensitivity to activated complement based on their differentiation and culture conditions. In organotypic cultures, the protection of neurons, OLs, and OPCs against CDC is eliminated by targeted astrocyte destruction. The activation of human complement proteins on mouse CNS cells necessitates caution when interpreting the results of mouse experimental models of NMO using HC. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0767-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Danielle E Harlow
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Katherine S Given
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Gregory P Owens
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Wendy B Macklin
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA.,Program in Neuroscience, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Jeffrey L Bennett
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA. .,Department of Ophthalmology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA. .,Program in Neuroscience, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA.
| |
Collapse
|
42
|
Shi K, Wang Z, Liu Y, Gong Y, Fu Y, Li S, Wood K, Hao J, Zhang GX, Shi FD, Yan Y. CFHR1-Modified Neural Stem Cells Ameliorated Brain Injury in a Mouse Model of Neuromyelitis Optica Spectrum Disorders. THE JOURNAL OF IMMUNOLOGY 2016; 197:3471-3480. [DOI: 10.4049/jimmunol.1600135] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 08/25/2016] [Indexed: 01/19/2023]
|
43
|
Kim JS, Cheon S, Kim SW, Kim B, Kim H, Park KD, Kim SM. Glycyrrhizic acid prevents astrocyte death by neuromyelitis optica-specific IgG via inhibition of C1q binding. Biochem Biophys Res Commun 2016; 478:553-8. [DOI: 10.1016/j.bbrc.2016.07.098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/21/2016] [Indexed: 11/15/2022]
|
44
|
Abstract
The complement system is a major component of innate immunity and a potent driver of inflammation. It has key roles in host defense against pathogens but can also contribute to pathology by driving inflammation and cell damage in diverse diseases. Complement has emerged as an important factor in the pathogenesis of numerous diseases of the CNS and PNS, including infectious, autoimmune and degenerative disorders, and is increasingly implicated in neuropsychiatric disease. Establishing the roles and relevance of complement in disease pathogenesis has become ever more important in recent years as new drugs targeting the complement system have reached the clinic, and the potential for using complement analytes as disease biomarkers has been recognized. In this brief review, the author summarizes the evidence implicating complement in these diseases and outlines ways in which this new understanding can be used to aid diagnosis and improve outcome.
Collapse
Affiliation(s)
- Bryan Paul Morgan
- a Institute of Infection and Immunity, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff CF144XN, UK
| |
Collapse
|
45
|
Yao X, Su T, Verkman AS. Clobetasol promotes remyelination in a mouse model of neuromyelitis optica. Acta Neuropathol Commun 2016; 4:42. [PMID: 27117475 PMCID: PMC4845317 DOI: 10.1186/s40478-016-0309-4] [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: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 12/14/2022] Open
Abstract
Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system that can produce marked neurological deficit. Current NMO therapies include immunosuppressants, plasma exchange and B-cell depletion. Here, we evaluated 14 potential remyelinating drugs emerging from prior small molecule screens done to identify drugs for repurposing in multiple sclerosis and other demyelinating neurological diseases. Compounds were initially evaluated in oligodendrocyte precursor cell (OPC) and cerebellar slice cultures, and then in a mouse model of NMO produced by intracerebral injection of anti-AQP4 autoantibody (AQP4-IgG) and human complement characterized by demyelination with minimal axonal damage. The FDA-approved drug clobetasol promoted differentiation in OPC cultures and remyelination in cerebellar slice cultures and in mice. Intraperitoneal administration of 2 mg/kg/day clobetasol reduced myelin loss by ~60 %, even when clobetasol was administered after demyelination occurred. Clobetasol increased the number of mature oligodendrocytes within lesions without significantly altering initial astrocyte damage or inflammation. These results provide proof-of-concept for the potential utility of a remyelinating approach in the treatment of NMO.
Collapse
|
46
|
McGonigal R, Cunningham ME, Yao D, Barrie JA, Sankaranarayanan S, Fewou SN, Furukawa K, Yednock TA, Willison HJ. C1q-targeted inhibition of the classical complement pathway prevents injury in a novel mouse model of acute motor axonal neuropathy. Acta Neuropathol Commun 2016; 4:23. [PMID: 26936605 PMCID: PMC4776408 DOI: 10.1186/s40478-016-0291-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/18/2023] Open
Abstract
Introduction Guillain-Barré syndrome (GBS) is an autoimmune disease that results in acute paralysis through inflammatory attack on peripheral nerves, and currently has limited, non-specific treatment options. The pathogenesis of the acute motor axonal neuropathy (AMAN) variant is mediated by complement-fixing anti-ganglioside antibodies that directly bind and injure the axon at sites of vulnerability such as nodes of Ranvier and nerve terminals. Consequently, the complement cascade is an attractive target to reduce disease severity. Recently, C5 complement component inhibitors that block the formation of the membrane attack complex and subsequent downstream injury have been shown to be efficacious in an in vivo anti-GQ1b antibody-mediated mouse model of the GBS variant Miller Fisher syndrome (MFS). However, since gangliosides are widely expressed in neurons and glial cells, injury in this model was not targeted exclusively to the axon and there are currently no pure mouse models for AMAN. Additionally, C5 inhibition does not prevent the production of early complement fragments such as C3a and C3b that can be deleterious via their known role in immune cell and macrophage recruitment to sites of neuronal damage. Results and Conclusions In this study, we first developed a new in vivo transgenic mouse model of AMAN using mice that express complex gangliosides exclusively in neurons, thereby enabling specific targeting of axons with anti-ganglioside antibodies. Secondly, we have evaluated the efficacy of a novel anti-C1q antibody (M1) that blocks initiation of the classical complement cascade, in both the newly developed anti-GM1 antibody-mediated AMAN model and our established MFS model in vivo. Anti-C1q monoclonal antibody treatment attenuated complement cascade activation and deposition, reduced immune cell recruitment and axonal injury, in both mouse models of GBS, along with improvement in respiratory function. These results demonstrate that neutralising C1q function attenuates injury with a consequent neuroprotective effect in acute GBS models and promises to be a useful new target for human therapy.
Collapse
|
47
|
Hinson SR, Lennon VA, Pittock SJ. Autoimmune AQP4 channelopathies and neuromyelitis optica spectrum disorders. HANDBOOK OF CLINICAL NEUROLOGY 2016; 133:377-403. [PMID: 27112688 DOI: 10.1016/b978-0-444-63432-0.00021-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neuromyelitis optica (NMO) spectrum disorders (SD) represent an evolving group of central nervous system (CNS)-inflammatory autoimmune demyelinating diseases unified by a pathogenic autoantibody specific for the aquaporin-4 (AQP4) water channel. It was historically misdiagnosed as multiple sclerosis (MS), which lacks a distinguishing biomarker. The discovery of AQP4-IgG moved the focus of CNS demyelinating disease research from emphasis on the oligodendrocyte and myelin to the astrocyte. NMO is recognized today as a relapsing disease, extending beyond the optic nerves and spinal cord to include brain (especially in children) and skeletal muscle. Brain magnetic resonance imaging abnormalities, identifiable in 60% of patients at the second attack, are consistent with MS in 10% of cases. NMOSD-typical lesions (another 10%) occur in AQP4-enriched regions: circumventricular organs (causing intractable nausea and vomiting) and the diencephalon (causing sleep disorders, endocrinopathies, and syndrome of inappropriate antidiuresis). Advances in understanding the immunobiology of AQP4 autoimmunity have necessitated continuing revision of NMOSD clinical diagnostic criteria. Assays that selectively detect pathogenic AQP4-IgG targeting extracellular epitopes of AQP4 are promising prognostically. When referring to AQP4 autoimmunity, we suggest substituting the term "autoimmune aquaporin-4 channelopathy" for the term "NMO spectrum disorders." Randomized clinical trials are currently assessing the efficacy and safety of newer immunotherapies. Increasing therapeutic options based on understanding the molecular pathogenesis is anticipated to improve the outcome for patients with AQP4 channelopathy.
Collapse
Affiliation(s)
- Shannon R Hinson
- Departments of Laboratory Medicine/Pathology and Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Vanda A Lennon
- Departments of Laboratory Medicine/Pathology and Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Sean J Pittock
- Departments of Laboratory Medicine/Pathology and Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA.
| |
Collapse
|
48
|
Comprehensive Characterization of Relationship Between Higher-Order Structure and FcRn Binding Affinity of Stress-Exposed Monoclonal Antibodies. Pharm Res 2015; 33:994-1002. [PMID: 26694753 DOI: 10.1007/s11095-015-1845-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/10/2015] [Indexed: 02/01/2023]
Abstract
PURPOSE In biopharmaceutical development, information regarding higher-order structure (HOS) is important to verify quality and characterize protein derivatives. In this study, we aimed to characterize the association between HOS and pharmacokinetic property of a stress-exposed monoclonal antibody (mAb). METHODS Purity, primary structure, thermal stability, and HOS were evaluated for mAbs exposed to heat, photo-irradiation, and chemical oxidation. To investigate conformation of stress-exposed mAbs, hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) was utilized. RESULTS No distinct difference in secondary or tertiary structure between stress-exposed and non-stressed samples was found by conventional spectroscopic techniques. In binding activity with the neonatal Fc receptor (FcRn), however, a marked decline was observed for force-oxidized mAb and a slight decline was observed for heat- and photodegraded mAbs. Using differential scanning calorimetry, a change in thermal stability was observed in the CH2 domain for all the stress-exposed samples. Using HDX-MS analyses, individual regions with altered conformation could be identified for heat-degraded and force-oxidized samples. CONCLUSIONS These findings indicate that comprehensive study is important for detecting conformational changes and helpful for predicting biophysical property, and that the evaluation of HOS using several analytical techniques is indispensable for confirming biopharmaceutical quality.
Collapse
|
49
|
Abstract
Complement is a key component of immunity with crucial inflammatory and opsonic properties; inappropriate activation of complement triggers or exacerbates inflammatory disease. Complement dysregulation is a core feature of some diseases and contributes to pathology in many others. Approved agents have been developed for and are highly effective in some orphan applications, but their progress to use in more common diseases has been slow. Numerous challenges, such as target concentration or high turnover, limit the efficacy of these agents in humans. Numerous novel agents targeting different parts of the complement system in different ways are now emerging from pre-clinical studies and are entering Phase I/II trials; these agents bring the potential for more-effective and more-specific anti-complement therapies in disease. Other agents, both biologic and small molecule, are in Phase II or III trials for both rare and common diseases — administration routes include localized (for example, intravitreal) and systemic routes. There is an urgent need to develop biomarkers and imaging methods that enable monitoring of the effects and efficacy of anti-complement agents.
The complement cascade, a key regulator of innate immunity, is a rich source of potential therapeutic targets for diseases including autoimmune, inflammatory and degenerative disorders. Morgan and Harris discuss the progress made in modulating the complement system and the existing challenges, including dosing, localization of the drug to the target and how to interfere with protein–protein interactions. The complement system is a key innate immune defence against infection and an important driver of inflammation; however, these very properties can also cause harm. Inappropriate or uncontrolled activation of complement can cause local and/or systemic inflammation, tissue damage and disease. Complement provides numerous options for drug development as it is a proteolytic cascade that involves nine specific proteases, unique multimolecular activation and lytic complexes, an arsenal of natural inhibitors, and numerous receptors that bind to activation fragments. Drug design is facilitated by the increasingly detailed structural understanding of the molecules involved in the complement system. Only two anti-complement drugs are currently on the market, but many more are being developed for diseases that include infectious, inflammatory, degenerative, traumatic and neoplastic disorders. In this Review, we describe the history, current landscape and future directions for anti-complement therapies.
Collapse
|
50
|
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
|