1
|
Yamamoto M, Itokazu T, Uno H, Maki T, Shibuya N, Yamashita T. Anti-RGMa neutralizing antibody ameliorates vascular cognitive impairment in mice. Neurotherapeutics 2025; 22:e00500. [PMID: 39613526 PMCID: PMC12014345 DOI: 10.1016/j.neurot.2024.e00500] [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: 07/12/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024] Open
Abstract
Repulsive Guidance Molecule A (RGMa) is well-recognized for its role in axon guidance. Recent studies have unveiled its diverse functions under pathological conditions within the central nervous system, such as spinal cord injury, multiple sclerosis, and Parkinson's disease. In this study, we explored the involvement of RGMa and the therapeutic effects of an anti-RGMa neutralizing antibody in a mouse model of vascular dementia (VaD). The VaD mouse model was established using the bilateral common carotid artery stenosis (BCAS) method. Immunohistochemical analysis revealed that these mice exhibited increased RGMa expression in the hippocampus, which coincided with reduced neurogenesis and impaired cholinergic innervation. These alterations manifested as cognitive impairments in the BCAS mice. Significantly, treatment with anti-RGMa neutralizing antibody reversed these pathological changes and cognitive deficits. Our findings suggest that RGMa plays a pivotal role in VaD pathology within the hippocampus and propose the anti-RGMa antibody as a promising therapeutic avenue for treating VaD.
Collapse
Affiliation(s)
- Masaya Yamamoto
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| | - Hiroki Uno
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nao Shibuya
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan; WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan.
| |
Collapse
|
2
|
Gerasimova E, Beenen AC, Kachkin D, Regensburger M, Zundler S, Blumenthal DB, Lutzny-Geier G, Winner B, Prots I. Novel co-culture model of T cells and midbrain organoids for investigating neurodegeneration in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:36. [PMID: 40021643 PMCID: PMC11871142 DOI: 10.1038/s41531-025-00882-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Recent studies demonstrate that brain infiltration of peripheral immune cells and their interaction with brain-resident cells contribute to Parkinson's disease (PD). However, mechanisms of T cell-brain cell communication are not fully elucidated and models allowing investigation of interaction between T cells and brain-resident cells are required. In this study, we developed a three-dimensional (3D) model composed of stem cell-derived human midbrain organoids (hMO) and peripheral blood T cells. We demonstrated that organoids consist of multiple midbrain-specific cell types, allowing to study T cell motility and interactions with midbrain tissue in a spatially organized microenvironment. We optimized co-culture conditions and demonstrated that T cells infiltrate hMO tissue, leading to neural cell loss. Our work establishes a novel 3D cell co-culture model as a promising tool to investigate the effect of the adaptive immune system on the midbrain and can be used in future studies to address these processes in the context of PD.
Collapse
Affiliation(s)
- Elizaveta Gerasimova
- Dental Clinic 1-Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Amke C Beenen
- Dental Clinic 1-Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniil Kachkin
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Regensburger
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Translational Research Center (TRC), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - David B Blumenthal
- Biomedical Network Science Lab, Department of Artificial Intelligence in Biomedical Engineering (AIBE), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gloria Lutzny-Geier
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Center for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Dental Clinic 1-Department of Operative Dentistry and Periodontology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
3
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2025; 36:139-168. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
4
|
Olejnik P, Roszkowska Z, Adamus S, Kasarełło K. Multiple sclerosis: a narrative overview of current pharmacotherapies and emerging treatment prospects. Pharmacol Rep 2024; 76:926-943. [PMID: 39177889 PMCID: PMC11387431 DOI: 10.1007/s43440-024-00642-0] [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: 06/13/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease characterized by pathological processes of demyelination, subsequent axonal loss, and neurodegeneration within the central nervous system. Despite the availability of numerous disease-modifying therapies that effectively manage this condition, there is an emerging need to identify novel therapeutic targets, particularly for progressive forms of MS. Based on contemporary insights into disease pathophysiology, ongoing efforts are directed toward developing innovative treatment modalities. Primarily, monoclonal antibodies have been extensively investigated for their efficacy in influencing specific pathological pathways not yet targeted. Emerging approaches emphasizing cellular mechanisms, such as chimeric antigen receptor T cell therapy targeting immunological cells, are attracting increasing interest. The evolving understanding of microglia and the involvement of ferroptotic mechanisms in MS pathogenesis presents further avenues for targeted therapies. Moreover, innovative treatment strategies extend beyond conventional approaches to encompass interventions that target alterations in microbiota composition and dietary modifications. These adjunctive therapies hold promise as complementary methods for the holistic management of MS. This narrative review aims to summarize current therapies and outline potential treatment methods for individuals with MS.
Collapse
Affiliation(s)
- Piotr Olejnik
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Roszkowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Sylwia Adamus
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
- Biomedical Physics Division, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Kaja Kasarełło
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
5
|
Bsteh G, Dal Bianco A, Zrzavy T, Berger T. Novel and Emerging Treatments to Target Pathophysiological Mechanisms in Various Phenotypes of Multiple Sclerosis. Pharmacol Rev 2024; 76:564-578. [PMID: 38719481 DOI: 10.1124/pharmrev.124.001073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/16/2024] Open
Abstract
The objective is to comprehensively review novel pharmacotherapies used in multiple sclerosis (MS) and the possibilities they may carry for therapeutic improvement. Specifically, we discuss pathophysiological mechanisms worth targeting in MS, ranging from well known targets, such as autoinflammation and demyelination, to more novel and advanced targets, such as neuroaxonal damage and repair. To set the stage, a brief overview of clinical MS phenotypes is provided, followed by a comprehensive recapitulation of both clinical and paraclinical outcomes available to assess the effectiveness of treatments in achieving these targets. Finally, we discuss various promising novel and emerging treatments, including their respective hypothesized modes of action and currently available evidence from clinical trials. SIGNIFICANCE STATEMENT: This comprehensive review discusses pathophysiological mechanisms worth targeting in multiple sclerosis. Various promising novel and emerging treatments, including their respective hypothesized modes of action and currently available evidence from clinical trials, are reviewed.
Collapse
Affiliation(s)
- Gabriel Bsteh
- Department of Neurology (G.B., A.D.B., T.Z., T.B.) and Comprehensive Center for Clinical Neurosciences & Mental Health (G.B., A.D.B., T.Z., T.B.), Medical University of Vienna, Vienna, Austria
| | - Assunta Dal Bianco
- Department of Neurology (G.B., A.D.B., T.Z., T.B.) and Comprehensive Center for Clinical Neurosciences & Mental Health (G.B., A.D.B., T.Z., T.B.), Medical University of Vienna, Vienna, Austria
| | - Tobias Zrzavy
- Department of Neurology (G.B., A.D.B., T.Z., T.B.) and Comprehensive Center for Clinical Neurosciences & Mental Health (G.B., A.D.B., T.Z., T.B.), Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology (G.B., A.D.B., T.Z., T.B.) and Comprehensive Center for Clinical Neurosciences & Mental Health (G.B., A.D.B., T.Z., T.B.), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Shimizu M, Shiraishi N, Tada S, Sasaki T, Beck G, Nagano S, Kinoshita M, Sumi H, Sugimoto T, Ishida Y, Koda T, Ishikura T, Sugiyama Y, Kihara K, Kanakura M, Nakajima T, Takeda S, Takahashi MP, Yamashita T, Okuno T, Mochizuki H. RGMa collapses the neuronal actin barrier against disease-implicated protein and exacerbates ALS. SCIENCE ADVANCES 2023; 9:eadg3193. [PMID: 37992159 PMCID: PMC10665002 DOI: 10.1126/sciadv.adg3193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 10/23/2023] [Indexed: 11/24/2023]
Abstract
Repulsive guidance molecule A (RGMa) was originally identified as a neuronal growth cone-collapsing factor. Previous reports have demonstrated the multifunctional roles of RGMa mediated by neogenin1. However, the pathogenic involvement of RGMa in amyotrophic lateral sclerosis (ALS) remains unclear. Here, we demonstrated that RGMa concentration was elevated in the cerebrospinal fluid of both patients with ALS and transgenic mice overexpressing the mutant human superoxide dismutase1 (mSOD1 mice). Treatment with humanized anti-RGMa monoclonal antibody ameliorated the clinical symptoms in mSOD1 mice. Histochemical analysis revealed that the anti-RGMa antibody significantly decreased mutant SOD1 protein accumulation in the motor neurons of mSOD1 mice via inhibition of actin depolymerization. In vitro analysis revealed that the anti-RGMa antibody inhibited the cellular uptake of the mutant SOD1 protein, presumably by reinforcing the neuronal actin barrier. Collectively, these data suggest that RGMa leads to the collapse of the neuronal actin barrier and promotes aberrant protein deposition, resulting in exacerbation of the ALS pathology.
Collapse
Affiliation(s)
- Mikito Shimizu
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Naoyuki Shiraishi
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoru Tada
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Clinical Research, National Hospital Organization Osaka-Minami Medical Center, Kawachinagano, Osaka, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Goichi Beck
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Seiichi Nagano
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Neurotherapeutics, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Makoto Kinoshita
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hisae Sumi
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Neurology, Higashiosaka City Medical Center, Higashiosaka, Osaka, Japan
| | - Tomoyuki Sugimoto
- Graduate School of Data Science, Shiga University, Hikone, Shiga, Japan
| | - Yoko Ishida
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Toru Koda
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Teruyuki Ishikura
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Neurology, Higashiosaka City Medical Center, Higashiosaka, Osaka, Japan
| | - Yasuko Sugiyama
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keigo Kihara
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Minami Kanakura
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Health Sciences, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tsuneo Nakajima
- Department of Geriatric and General Medicine, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Hirakata, Osaka, Japan
| | - Masanori P. Takahashi
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Health Sciences, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
7
|
Mothe AJ, Jacobson PB, Caprelli M, Ulndreaj A, Rahemipour R, Huang L, Monnier PP, Fehlings MG, Tator CH. Delayed administration of elezanumab, a human anti-RGMa neutralizing monoclonal antibody, promotes recovery following cervical spinal cord injury. Neurobiol Dis 2022; 172:105812. [PMID: 35810963 DOI: 10.1016/j.nbd.2022.105812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) elicits a cascade of degenerative events including cell death, axonal degeneration, and the upregulation of inhibitory molecules which limit repair. Repulsive guidance molecule A (RGMa) is an axon growth inhibitor which is also involved in neuronal cell death and differentiation. SCI causes upregulation of RGMa in the injured rodent, non-human primate, and human spinal cord. Recently, we showed that delayed administration of elezanumab, a high affinity human RGMa-specific monoclonal antibody, promoted neuroprotective and regenerative effects following thoracic SCI. Since most human traumatic SCI is at the cervical level, and level-dependent anatomical and molecular differences may influence pathophysiological responses to injury and treatment, we examined the efficacy of elezanumab and its therapeutic time window of administration in a clinically relevant rat model of cervical impact-compression SCI. Pharmacokinetic analysis of plasma and spinal cord tissue lysate showed comparable levels of RGMa antibodies with delayed administration following cervical SCI. At 12w after SCI, elezanumab promoted long term benefits including perilesional sparing of motoneurons and increased neuroplasticity of key descending pathways involved in locomotion and fine motor function. Elezanumab also promoted growth of corticospinal axons into spinal cord gray matter and enhanced serotonergic innervation of the ventral horn to form synaptic connections caudal to the cervical lesion. Significant recovery in grip and trunk/core strength, locomotion and gait, and spontaneous voiding ability was found in rats treated with elezanumab either immediately post-injury or at 3 h post-SCI, and improvements in specific gait parameters were found when elezanumab was delayed to 24 h post-injury. We also developed a new locomotor score, the Cervical Locomotor Score, a simple and sensitive measure of trunk/core and limb strength and stability during dynamic locomotion.
Collapse
Affiliation(s)
- Andrea J Mothe
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada.
| | - Peer B Jacobson
- Department of Translational Sciences, AbbVie Inc., North Chicago, IL 60064, USA
| | - Mitchell Caprelli
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Antigona Ulndreaj
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Radmehr Rahemipour
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada
| | - Lili Huang
- AbbVie Biologics, AbbVie Bioresearch Center, Worcester, MA 01605, USA
| | - Philippe P Monnier
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, M5S 3H6, ON, Canada
| | - Michael G Fehlings
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, M5T 2S8, ON, Canada
| | - Charles H Tator
- Division of Experimental and Translational Neuroscience, Krembil Brain Institute & University Health Network, Toronto, M5T 0S8, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, M5T 2S8, ON, Canada.
| |
Collapse
|
8
|
Yu T, Huo L, Lei J, Sun JJ, Wang H. Modulation of Microglia M2 Polarization and Alleviation of Hippocampal Neuron Injury By MiR-106b-5p/RGMa in a Mouse Model of Status Epilepticus. Inflammation 2022; 45:2223-2242. [PMID: 35789312 DOI: 10.1007/s10753-022-01686-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022]
Abstract
MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level. The miRNA miR-106b-5p has been linked to epilepsy, but its specific role and mechanism of action remain unclear. This was investigated in the present study using a mouse model of pilocarpine-induced status epilepticus and an in vitro system of HT22 hippocampal cells treated with Mg2+-free solution and cocultured with BV2 microglia cells. We found that inhibiting miR-106b-5p expression promoted microglia M2 polarization, reduced the inflammatory response, and alleviated neuronal injury. These effects involved modulation of the repulsive guidance molecule A (RGMa)-Rac1-c-Jun N-terminal kinase (JNK)/p38-mitogen-activated protein kinase (MAPK) signaling axis. Our results suggest that therapeutic strategies targeting miR-106b-5p or downstream factors can be effective in preventing epileptogenesis or treating epilepsy.
Collapse
Affiliation(s)
- Tao Yu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang City, 110004, China
| | - Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang City, 110004, China
| | - Jie Lei
- Department of Pediatrics, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang City, 110004, China
| | - Jing-Jing Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang City, 110004, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Liaoning Province, Shenyang City, 110004, China.
| |
Collapse
|
9
|
Iwamoto S, Itokazu T, Sasaki A, Kataoka H, Tanaka S, Hirata T, Miwa K, Suenaga T, Takai Y, Misu T, Fujihara K, Yamashita T. RGMa signal in Macrophages Induces Neutrophil-related Astrocytopathy in NMO. Ann Neurol 2022; 91:532-547. [PMID: 35167145 DOI: 10.1002/ana.26327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Repulsive guidance molecule-a (RGMa) is a glycosylphosphatidylinositol-linked glycoprotein which has multiple functions including axon growth inhibition and immune regulation. However, its role in the pathophysiology of neuromyelitis optica (NMO) is poorly understood. Perivascular astrocytopathy, which is induced by the leakage of aquaporin-4 (AQP4)-specific IgG into the central nervous system parenchyma, is a key feature of NMO pathology. We investigated the RGMa involvement in the pathology of NMO astrocytopathy, and tested a therapeutic potential of humanized anti-RGMa monoclonal antibody (RGMa-mAb). METHODS Using a clinically relevant NMO rat model, we evaluated the therapeutic effect of a RGMa-mAb by behavioral testing, immunohistochemistry, and gene expression assay. We further performed in vitro experiments to address the RGMa-signaling in macrophages. RESULTS In both NMO rats and an NMO-autopsied sample, RGMa was expressed by the spared neurons and astrocytes, whereas its receptor neogenin was expressed by infiltrating macrophages. AQP4-IgG-induced astrocytopathy and clinical exacerbation in NMO rats were ameliorated by RGMa-mAb treatment. RGMa-mAb treatment significantly suppressed neutrophil infiltration, and decreased the expression of neutrophil chemoattractants. Interestingly, neogenin-expressing macrophages accumulated in the lesion expressed CXCL2, a strong neutrophil chemoattractant, and further analysis revealed that RGMa directly regulated CXCL2 expression in macrophages. Finally, we found that our NMO rats developed neuropathic pain, and RGMa-mAb treatment effectively ameliorated the severity of neuropathic pain. INTERPRETATION RGMa signaling in infiltrated macrophages is a critical driver of neutrophil-related astrocytopathy in NMO lesions, and RGMa-mAb may provide an efficient therapeutic strategy for NMO-associated neuropathic pain and motor deficits in patients with NMO. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shosuke Iwamoto
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Takahide Itokazu
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Molecular Neurosciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Atsushi Sasaki
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Hirotoshi Kataoka
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Shinji Tanaka
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Takeshi Hirata
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | - Keiko Miwa
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | | | - Yoshiki Takai
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuro Misu
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuo Fujihara
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshihide Yamashita
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Molecular Neurosciences, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI-Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
10
|
Tang J, Zeng X, Yang J, Zhang L, Li H, Chen R, Tang S, Luo Y, Qin X, Feng J. Expression and Clinical Correlation Analysis Between Repulsive Guidance Molecule a and Neuromyelitis Optica Spectrum Disorders. Front Immunol 2022; 13:766099. [PMID: 35185873 PMCID: PMC8850277 DOI: 10.3389/fimmu.2022.766099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives This study sought to explore the expression patterns of repulsive guidance molecules a (RGMa) in neuromyelitis optica spectrum disorders (NMOSD) and to explore the correlation between RGMa and the clinical features of NMOSD. Methods A total of 83 NMOSD patients and 22 age-matched healthy controls (HCs) were enrolled in the study from October 2017 to November 2021. Clinical parameters, including Expanded Disability Status Scale (EDSS) score, degree of MRI enhancement, and AQP4 titer were collected. The expression of serum RGMa was measured by enzyme-linked immunosorbent assay (ELISA) and compared across the four patient groups. The correlation between serum RGMa levels and different clinical parameters was also assessed. Results The average serum expression of RGMa in the NMOSD group was significantly higher than that in the HC group (p < 0.001). Among the patient groups, the acute phase group exhibited significantly higher serum RGMa levels than did the remission group (p < 0.001). A multivariate analysis revealed a significant positive correlation between RGMa expression and EDSS score at admission, degree of MRI enhancement, and segmental length of spinal cord lesions. There was a significant negative correlation between the expression of RGMa in NMOSD and the time from attack to sampling or delta EDSS. Conclusions The current study suggests that RGMa may be considered a potential biomarker predicting the severity, disability, and clinical features of NMOSD.
Collapse
Affiliation(s)
- Jinhua Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, People's Hospital of Chongqing Hechuan, Chongqing, China
| | - Xiaopeng Zeng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hang Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shi Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yetao Luo
- Department of Biostatistics, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Huang L, Fung E, Bose S, Popp A, Böser P, Memmott J, Kutskova YA, Miller R, Tarcsa E, Klein C, Veldman GM, Mueller BK, Cui YF. Elezanumab, a clinical stage human monoclonal antibody that selectively targets repulsive guidance molecule A to promote neuroregeneration and neuroprotection in neuronal injury and demyelination models. Neurobiol Dis 2021; 159:105492. [PMID: 34478849 DOI: 10.1016/j.nbd.2021.105492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022] Open
Abstract
Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lili Huang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Emma Fung
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Sahana Bose
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Andreas Popp
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - Preethne Böser
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - John Memmott
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Yuliya A Kutskova
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Renee Miller
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Edit Tarcsa
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Corinna Klein
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | | | - Bernhard K Mueller
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - Yi-Fang Cui
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| |
Collapse
|
12
|
Yan XZ, Lai L, Ao Q, Tian XH, Zhang YH. Interleukin-17A in Alzheimer's disease: recent advances and controversies. Curr Neuropharmacol 2021; 20:372-383. [PMID: 34429057 PMCID: PMC9413786 DOI: 10.2174/1570159x19666210823110004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/03/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that mainly affects older adults. Although the global burden of AD is increasing year by year, the causes of AD remain largely unknown. Numerous basic and clinical studies have shown that interleukin-17A (IL-17A) may play a significant role in the pathogenesis of AD. A comprehensive assessment of the role of IL-17A in AD would benefit the diagnosis, understanding of etiology and treatment. However, over the past decade, controversies remain regarding the expression level and role of IL-17A in AD. We have incorporated newly published researches and point out that IL-17A expression levels may vary along with the development of AD, exercising different roles at different stages of AD, although much more work remains to be done to support the potential role of IL-17A in AD-related pathology. Here, it is our intention to review the underlying mechanisms of IL-17A in AD and address the current controversies in an effort to clarify the results of existing research and suggest future studies.
Collapse
Affiliation(s)
- Xin-Zhu Yan
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT. 0
| | - Qiang Ao
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064. China
| | - Xiao-Hong Tian
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| | - Yan-Hui Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| |
Collapse
|
13
|
Müller T. View Point: Disease Modification and Cell Secretome Based Approaches in Parkinson's Disease: Are We on the Right Track? Biologics 2021; 15:307-316. [PMID: 34349499 PMCID: PMC8328382 DOI: 10.2147/btt.s267281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022]
Abstract
The term idiopathic Parkinson's disease describes an entity of various not well-characterized disorders resembling each other. They are characterized by chronic neuronal dying originating from various disease mechanisms. They result in the onset of motor and related non-motor features, both of which respond to administration of personalized drug combinations and surgical therapies. The unmet need is beneficial disease course modification with repair and neurogenesis. Objectives are to discuss the value of cell secretome based treatments including neuronal graft transplantation and to suggest as an alternative the stimulation of an endogenous available approach for neuronal repair. Chronic neurodegenerative processes result from different heterogeneous, but complementing metabolic, pathological cascade sequences. Accumulated evidence from experimental research suggested neuron transplantation, stem cell application and cell secretome-based therapies as a promising future treatment with cure as an ultimate goal. To date, clinical testing of disease-modifying treatments has focused on substitution or repair of the remaining dopamine synthesizing neurons following diagnosis. At diagnosis, many of the still surviving and functioning, but already affected neurons have lost most of their axons and are primed for cell death. A more promising therapeutic concept may be the stimulation of an existing, endogenous repair system in the peripheral and central nervous systems. The abundant protein repulsive guidance molecule A blocks restoration and neurogenesis, both of which are mediated via the neogenin receptor. Inhibition of the physiological effects of repulsive guidance molecule A is an endogenous available repair pathway in chronic neurodegeneration. Antagonism of this protein with antibodies or stimulation of the neogenin receptor should be considered as an initial repair step. It is an alternative to cell replacement, stem cell or associated cell secretome concepts.
Collapse
Affiliation(s)
- Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weissensee, Berlin, 13088, Germany
| |
Collapse
|
14
|
Sutiwisesak R, Burns TC, Rodriguez M, Warrington AE. Remyelination therapies for multiple sclerosis: optimizing translation from animal models into clinical trials. Expert Opin Investig Drugs 2021; 30:857-876. [PMID: 34126015 DOI: 10.1080/13543784.2021.1942840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Multiple sclerosis (MS) is the most common inflammatory disease of the central nervous system (CNS). Demyelination, the main pathology in MS, contributes to clinical symptoms and long-term neurological deficits if left untreated. Remyelination, the natural repair of damaged myelin by cells of the oligodendrocyte lineage, occurs in MS, but eventually fails in most patients as they age. Encouraging timely remyelination can restore axon conduction and minimize deficits.Areas covered: We discuss and correlate human MS pathology with animal models, propose methods to deplete resident oligodendrocyte progenitor cells (OPCs) to determine whether mature oligodendrocytes support remyelination, and review remyelinating agents, mechanisms of action, and available clinical trial data.Expert opinion: The heterogeneity of human MS may limit successful translation of many candidate remyelinating agents; some patients lack the biological targets necessary to leverage current approaches. Development of therapeutics for remyelination has concentrated almost exclusively on mobilization of innate OPCs. However, mature oligodendrocytes appear an important contributor to remyelination in humans. Limiting the contribution of OPC mediated repair in models of MS would allow the evaluation of remyelination-promoting agents on mature oligodendrocytes. Among remyelinating reagents reviewed, only rHIgM22 targets both OPCs and mature oligodendrocytes.
Collapse
Affiliation(s)
- Rujapope Sutiwisesak
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Terry C Burns
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| | - Moses Rodriguez
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur E Warrington
- Departments of Neurology and Neurologic Surgery Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Repulsive Guidance Molecule-a and Central Nervous System Diseases. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5532116. [PMID: 33997000 PMCID: PMC8112912 DOI: 10.1155/2021/5532116] [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: 02/05/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Repulsive guidance molecule-a (RGMa) is a member of glycosylphosphatidylinositol- (GPI-) anchored protein family, which has axon guidance function and is widely involved in the development and pathological processes of the central nervous system (CNS). On the one hand, the binding of RGMa and its receptor Neogenin can regulate axonal guidance, differentiation of neural stem cells into neurons, and the survival of these cells; on the other hand, RGMa can inhibit functional recovery of CNS by inhibiting axonal growth. A number of studies have shown that RGMa may be involved in the pathogenesis of CNS diseases, such as multiple sclerosis, neuromyelitis optica spectrum diseases, cerebral infarction, spinal cord injury, Parkinson's disease, and epilepsy. Targeting RGMa can enhance the functional recovery of CNS, so it may become a promising target for the treatment of CNS diseases. This article will comprehensively review the research progression of RGMa in various CNS diseases up to date.
Collapse
|
16
|
Perspective: Treatment for Disease Modification in Chronic Neurodegeneration. Cells 2021; 10:cells10040873. [PMID: 33921342 PMCID: PMC8069143 DOI: 10.3390/cells10040873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Symptomatic treatments are available for Parkinson's disease and Alzheimer's disease. An unmet need is cure or disease modification. This review discusses possible reasons for negative clinical study outcomes on disease modification following promising positive findings from experimental research. It scrutinizes current research paradigms for disease modification with antibodies against pathological protein enrichment, such as α-synuclein, amyloid or tau, based on post mortem findings. Instead a more uniform regenerative and reparative therapeutic approach for chronic neurodegenerative disease entities is proposed with stimulation of an endogenously existing repair system, which acts independent of specific disease mechanisms. The repulsive guidance molecule A pathway is involved in the regulation of peripheral and central neuronal restoration. Therapeutic antagonism of repulsive guidance molecule A reverses neurodegeneration according to experimental outcomes in numerous disease models in rodents and monkeys. Antibodies against repulsive guidance molecule A exist. First clinical studies in neurological conditions with an acute onset are under way. Future clinical trials with these antibodies should initially focus on well characterized uniform cohorts of patients. The efficiency of repulsive guidance molecule A antagonism and associated stimulation of neurogenesis should be demonstrated with objective assessment tools to counteract dilution of therapeutic effects by subjectivity and heterogeneity of chronic disease entities. Such a research concept will hopefully enhance clinical test strategies and improve the future therapeutic armamentarium for chronic neurodegeneration.
Collapse
|
17
|
Molecular Mechanisms of Central Nervous System Axonal Regeneration and Remyelination: A Review. Int J Mol Sci 2020; 21:ijms21218116. [PMID: 33143194 PMCID: PMC7662268 DOI: 10.3390/ijms21218116] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Central nervous system (CNS) injury, including stroke, spinal cord injury, and traumatic brain injury, causes severe neurological symptoms such as sensory and motor deficits. Currently, there is no effective therapeutic method to restore neurological function because the adult CNS has limited capacity to regenerate after injury. Many efforts have been made to understand the molecular and cellular mechanisms underlying CNS regeneration and to establish novel therapeutic methods based on these mechanisms, with a variety of strategies including cell transplantation, modulation of cell intrinsic molecular mechanisms, and therapeutic targeting of the pathological nature of the extracellular environment in CNS injury. In this review, we will focus on the mechanisms that regulate CNS regeneration, highlighting the history, recent efforts, and questions left unanswered in this field.
Collapse
|
18
|
Liu F, Cheng X, Zhong S, Liu C, Jolkkonen J, Zhang X, Liang Y, Liu Z, Zhao C. Communications Between Peripheral and the Brain-Resident Immune System in Neuronal Regeneration After Stroke. Front Immunol 2020; 11:1931. [PMID: 33042113 PMCID: PMC7530165 DOI: 10.3389/fimmu.2020.01931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/17/2020] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemia may cause irreversible neural network damage and result in functional deficits. Targeting neuronal repair after stroke potentiates the formation of new connections, which can be translated into a better functional outcome. Innate and adaptive immune responses in the brain and the periphery triggered by ischemic damage participate in regulating neural repair after a stroke. Immune cells in the blood circulation and gut lymphatic tissues that have been shaped by immune components including gut microbiota and metabolites can infiltrate the ischemic brain and, once there, influence neuronal regeneration either directly or by modulating the properties of brain-resident immune cells. Immune-related signalings and metabolites from the gut microbiota can also directly alter the phenotypes of resident immune cells to promote neuronal regeneration. In this review, we discuss several potential mechanisms through which peripheral and brain-resident immune components can cooperate to promote first the resolution of neuroinflammation and subsequently to improved neural regeneration and a better functional recovery. We propose that new insights into discovery of regulators targeting pro-regenerative process in this complex neuro-immune network may lead to novel strategies for neuronal regeneration.
Collapse
Affiliation(s)
- Fangxi Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Xi Cheng
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Shanshan Zhong
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Jukka Jolkkonen
- A.I. Virtanen Institute and Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Xiuchun Zhang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Yifan Liang
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Zhouyang Liu
- Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, Shenyang, China.,Stroke Center, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
A Repulsive Environment Induces Neurodegeneration of Midbrain Dopaminergic Neurons. J Neurosci 2019; 38:1323-1325. [PMID: 29438033 DOI: 10.1523/jneurosci.3070-17.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/18/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022] Open
|
20
|
Chen J, Shifman MI. Inhibition of neogenin promotes neuronal survival and improved behavior recovery after spinal cord injury. Neuroscience 2019; 408:430-447. [PMID: 30943435 DOI: 10.1016/j.neuroscience.2019.03.055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/09/2023]
Abstract
Following spinal cord trauma, axonal regeneration in the mammalian spinal cord does not occur and functional recovery may be further impeded by retrograde neuronal death. By contrast, lampreys recover after spinal cord injury (SCI) and axons re-connected to their targets in spinal cord. However, the identified reticulospinal (RS) neurons located in the lamprey brain differ in their regenerative capacities - some are good regenerators, and others are bad regenerators - despite the fact that they have analogous projection pathways. Previously, we reported that axonal guidance receptor Neogenin involved in regulation of axonal regeneration after SCI and downregulation of Neogenin synthesis by morpholino oligonucleotides (MO) enhanced the regeneration of RS neurons. Incidentally, the bad regenerating RS neurons often undergo a late retrograde apoptosis after SCI. Here we report that, after SCI, expression of RGMa mRNA was upregulated around the transection site, while its receptor Neogenin continued to be synthesized almost inclusively in the "bad-regenerating" RS neurons. Inhibition of Neogenin by MO prohibited activation of caspases and improved the survival of RS neurons at 10 weeks after SCI. These data provide new evidence in vivo that Neogenin is involved in retrograde neuronal death and failure of axonal regeneration after SCI.
Collapse
Affiliation(s)
- Jie Chen
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Philadelphia, PA 19140, USA
| | - Michael I Shifman
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Philadelphia, PA 19140, USA; Department of Neuroscience, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
21
|
Rotwein P. Variation in the repulsive guidance molecule family in human populations. Physiol Rep 2019; 7:e13959. [PMID: 30746893 PMCID: PMC6370684 DOI: 10.14814/phy2.13959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 01/17/2023] Open
Abstract
Repulsive guidance molecules, RGMA, RGMB, and RGMC, are related proteins discovered independently through different experimental paradigms. They are encoded by single copy genes in mammalian and other vertebrate genomes, and are ~50% identical in amino acid sequence. The importance of RGM actions in human physiology has not been realized, as most research has focused on non-human models, although mutations in RGMC are the cause of the severe iron storage disorder, juvenile hemochromatosis. Here I show that repositories of human genomic and population genetic data can be used as starting points for discovery and for developing new testable hypotheses about each of these paralogs in human biology and disease susceptibility. Information was extracted, aggregated, and analyzed from the Ensembl and UCSC Genome Browsers, the Exome Aggregation Consortium, the Genotype-Tissue Expression project portal, the cBio portal for Cancer Genomics, and the National Cancer Institute Genomic Data Commons data site. Results identify extensive variation in gene expression patterns, substantial alternative RNA splicing, and possible missense alterations and other modifications in the coding regions of each of the three genes, with many putative mutations being detected in individuals with different types of cancers. Moreover, selected amino acid substitutions are highly prevalent in the world population, with minor allele frequencies of up to 37% for RGMA and up to 8% for RGMB. These results indicate that protein sequence variation is common in the human RGM family, and raises the possibility that individual variants will have a significant population impact on human physiology and/or disease predisposition.
Collapse
Affiliation(s)
- Peter Rotwein
- Department of Biomedical SciencesPaul L. Foster School of MedicineTexas Tech Health University Health Sciences CenterEl PasoTexas
| |
Collapse
|
22
|
Kant R, Pasi S, Surolia A. Auto-Reactive Th17-Cells Trigger Obsessive-Compulsive-Disorder Like Behavior in Mice With Experimental Autoimmune Encephalomyelitis. Front Immunol 2018; 9:2508. [PMID: 30429853 PMCID: PMC6220041 DOI: 10.3389/fimmu.2018.02508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/10/2018] [Indexed: 01/20/2023] Open
Abstract
Th17-lymphocytes are well known for their deleterious role in autoimmunity. But does the notoriety of this repertoire extend beyond autoimmunity? In the present study we employed experimental autoimmune encephalomyelitis as model system to study the role auto-reactive Th17 cells in neuropsychiatric disorders. The mice with experimental autoimmune encephalomyelitis exhibited exaggerated grooming activity. The observed behavioral anomaly resembled obsessive compulsive disorder (OCD) upon analysis of grooming microstructure, induced grooming, marble burying and nestlet shredding. The observed OCD like behavior was relieved upon Th17 cell depletion; alternatively, it could alone be induced by adoptive transfer of myelin oligodendrocyte glycoprotein (35-55) reactive Th17 in B6.Rag1−/− mice. The observed OCD like behavior was also alleviated upon treatment with a selective serotonin reuptake inhibitor, fluoxetine.
Collapse
Affiliation(s)
- Ravi Kant
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Shweta Pasi
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Avadhesha Surolia
- Molecular Science Laboratory, National Institute of Immunology, New Delhi, India.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| |
Collapse
|
23
|
Inhibiting repulsive guidance molecule-a suppresses secondary progression in mouse models of multiple sclerosis. Cell Death Dis 2018; 9:1061. [PMID: 30333477 PMCID: PMC6193044 DOI: 10.1038/s41419-018-1118-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/29/2018] [Accepted: 10/01/2018] [Indexed: 12/27/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system that is characterized by motor deficits, fatigue, pain, cognitive impairment, and sensory and visual dysfunction. Secondary progressive multiple sclerosis (SPMS) is a progressive form of MS that develops from relapsing-remitting MS. Repulsive guidance molecule-a (RGMa) has diverse functions, including axon growth inhibition and immune regulation. Here, we show inhibiting RGMa had therapeutic effects in mouse models of SPMS. We induced experimental autoimmune encephalomyelitis in nonobese diabetic mice (NOD-EAE mice) and treated them with humanized anti-RGMa monoclonal antibody. This treatment significantly suppressed secondary progression of disease and inflammation, demyelination and axonal degeneration. In addition, treatment with anti-RGMa antibody promoted the growth of corticospinal tracts and motor recovery in targeted EAE mice with inflammatory lesions in the spinal cord. Collectively, these results show that a humanized anti-RGMa antibody has therapeutic effects in mouse models of SPMS.
Collapse
|
24
|
RGMa mediates reactive astrogliosis and glial scar formation through TGFβ1/Smad2/3 signaling after stroke. Cell Death Differ 2018; 25:1503-1516. [PMID: 29396549 PMCID: PMC6113216 DOI: 10.1038/s41418-018-0058-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/11/2023] Open
Abstract
In response to stroke, astrocytes become reactive astrogliosis and are a major component of a glial scar. This results in the formation of both a physical and chemical (production of chondroitin sulfate proteoglycans) barrier, which prevent neurite regeneration that, in turn, interferes with functional recovery. However, the mechanisms of reactive astrogliosis and glial scar formation are poorly understood. In this work, we hypothesized that repulsive guidance molecule a (RGMa) regulate reactive astrogliosis and glial scar formation. We first found that RGMa was strongly expressed by reactive astrocytes in the glial scar in a rat model of middle cerebral artery occlusion/reperfusion. Genetic or pharmacologic inhibition of RGMa in vivo resulted in a strong reduction of reactive astrogliosis and glial scarring as well as in a pronounced improvement in functional recovery. Furthermore, we showed that transforming growth factor β1 (TGFβ1) stimulated RGMa expression through TGFβ1 receptor activin-like kinase 5 (ALK5) in primary cultured astrocytes. Knockdown of RGMa abrogated key steps of reactive astrogliosis and glial scar formation induced by TGFβ1, including cellular hypertrophy, glial fibrillary acidic protein upregulation, cell migration, and CSPGs secretion. Finally, we demonstrated that RGMa co-immunoprecipitated with ALK5 and Smad2/3. TGFβ1-induced ALK5-Smad2/3 interaction and subsequent phosphorylation of Smad2/3 were impaired by RGMa knockdown. Taken together, we identified that after stroke, RGMa promotes reactive astrogliosis and glial scar formation by forming a complex with ALK5 and Smad2/3 to promote ALK5-Smad2/3 interaction to facilitate TGFβ1/Smad2/3 signaling, thereby inhibiting neurological functional recovery. RGMa may be a new therapeutic target for stroke.
Collapse
|
25
|
Harada K, Fujita Y, Okuno T, Tanabe S, Koyama Y, Mochizuki H, Yamashita T. Inhibition of RGMa alleviates symptoms in a rat model of neuromyelitis optica. Sci Rep 2018; 8:34. [PMID: 29311561 PMCID: PMC5758562 DOI: 10.1038/s41598-017-18362-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/11/2017] [Indexed: 12/29/2022] Open
Abstract
Neuromyelitis optica (NMO) is an autoimmune disease associated with NMO immunoglobulin G (NMO-IgG), an antibody that selectively binds to the aquaporin-4. Here, we established a localized NMO model by injecting NMO-IgG into the spinal cord, and assessed the efficacy of treating its NMO-like symptoms by blocking repulsive guidance molecule-a (RGMa), an axon growth inhibitor. The model showed pathological features consistent with NMO. Systemic administration of humanized monoclonal anti-RGMa antibody delayed the onset and attenuated the severity of clinical symptoms. Further, it preserved astrocytes and reduced inflammatory-cell infiltration and axonal damage, suggesting that targeting RGMa is effective in treating NMO.
Collapse
Affiliation(s)
- Kana Harada
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Tatsusada Okuno
- Department of Immunopathology, Research Institute for Microbial Diseases, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.,Department of Neurology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shogo Tanabe
- WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
26
|
Sommer A, Winner B, Prots I. The Trojan horse - neuroinflammatory impact of T cells in neurodegenerative diseases. Mol Neurodegener 2017; 12:78. [PMID: 29078813 PMCID: PMC5658940 DOI: 10.1186/s13024-017-0222-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/19/2017] [Indexed: 11/27/2022] Open
Abstract
Neuronal degeneration is a common mechanism of many neurological diseases including Parkinson’s disease (PD), Alzheimer’s disease (AD), and Multiple Sclerosis (MS). While AD and PD are classical neurodegenerative diseases, the primary pathology in MS is driven by autoimmune inflammation, attacking oligodendrocytes and thereby inducing neurodegeneration. In AD and PD, immune cells are also considered to play an important role in the disease progression. While the role of local central nervous system (CNS) innate immune cells is well described, a potential influence of adaptive immune cells in PD and AD is not yet fully understood. Here, we aim to summarize findings concerning adaptive immune cells in PD pathogenesis and compare them to AD and MS. In the first part, we focus on disease-specific alterations of lymphocytes in the circulating blood. Subsequently, we describe what is known about CNS-infiltrated lymphocytes and mechanisms of their infiltration. Finally, we summarize published data and try to understand the mechanisms of how lymphocytes contribute to neurodegeneration in PD, AD, and MS. Lymphocytes are critically involved in the pathogenesis of MS, and clarifying the role of lymphocytes in PD and AD pathogenesis might lead to an identification of a common signature of lymphocytes in neurodegeneration and thus pave the road towards novel treatment options.
Collapse
Affiliation(s)
- Annika Sommer
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
27
|
Repulsive Guidance Molecule a (RGMa) Induces Neuropathological and Behavioral Changes That Closely Resemble Parkinson's Disease. J Neurosci 2017; 37:9361-9379. [PMID: 28842419 DOI: 10.1523/jneurosci.0084-17.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 07/12/2017] [Accepted: 08/11/2017] [Indexed: 01/06/2023] Open
Abstract
Repulsive guidance molecule member a (RGMa) is a membrane-associated or released guidance molecule that is involved in axon guidance, cell patterning, and cell survival. In our previous work, we showed that RGMa is significantly upregulated in the substantia nigra of patients with Parkinson's disease. Here we demonstrate the expression of RGMa in midbrain human dopaminergic (DA) neurons. To investigate whether RGMa might model aspects of the neuropathology of Parkinson's disease in mouse, we targeted RGMa to adult midbrain dopaminergic neurons using adeno-associated viral vectors. Overexpression of RGMa resulted in a progressive movement disorder, including motor coordination and imbalance, which is typical for a loss of DA release in the striatum. In line with this, RGMa induced selective degeneration of dopaminergic neurons in the substantia nigra (SN) and affected the integrity of the nigrostriatal system. The degeneration of dopaminergic neurons was accompanied by a strong microglia and astrocyte activation. The behavioral, molecular, and anatomical changes induced by RGMa in mice are remarkably similar to the clinical and neuropathological hallmarks of Parkinson's disease. Our data indicate that dysregulation of RGMa plays an important role in the pathology of Parkinson's disease, and antibody-mediated functional interference with RGMa may be a disease modifying treatment option.SIGNIFICANCE STATEMENT Parkinson's disease (PD) is a neurodegenerative disease characterized by severe motor dysfunction due to progressive degeneration of mesencephalic dopaminergic (DA) neurons in the substantia nigra. To date, there is no regenerative treatment available. We previously showed that repulsive guidance molecule member a (RGMa) is upregulated in the substantia nigra of PD patients. Adeno-associated virus-mediated targeting of RGMa to mouse DA neurons showed that overexpression of this repulsive axon guidance and cell patterning cue models the behavioral and neuropathological characteristics of PD in a remarkable way. These findings have implications for therapy development as interfering with the function of this specific axon guidance cue may be beneficial to the survival of DA neurons.
Collapse
|
28
|
Impact of aging immune system on neurodegeneration and potential immunotherapies. Prog Neurobiol 2017; 157:2-28. [PMID: 28782588 DOI: 10.1016/j.pneurobio.2017.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
The interaction between the nervous and immune systems during aging is an area of avid interest, but many aspects remain unclear. This is due, not only to the complexity of the aging process, but also to a mutual dependency and reciprocal causation of alterations and diseases between both the nervous and immune systems. Aging of the brain drives whole body systemic aging, including aging-related changes of the immune system. In turn, the immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution that are sources of chronic inflammation in the elderly (termed inflammaging), potentially induces brain aging and memory loss in a reciprocal manner. Therefore, immunotherapeutics including modulation of inflammation, vaccination, cellular immune therapies and "protective autoimmunity" provide promising approaches to rejuvenate neuroinflammatory disorders and repair brain injury. In this review, we summarize recent discoveries linking the aging immune system with the development of neurodegeneration. Additionally, we discuss potential rejuvenation strategies, focusing aimed at targeting the aging immune system in an effort to prevent acute brain injury and chronic neurodegeneration during aging.
Collapse
|
29
|
Localized Sympathectomy Reduces Mechanical Hypersensitivity by Restoring Normal Immune Homeostasis in Rat Models of Inflammatory Pain. J Neurosci 2017; 36:8712-25. [PMID: 27535916 DOI: 10.1523/jneurosci.4118-15.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 07/09/2016] [Indexed: 12/28/2022] Open
Abstract
UNLABELLED Some forms of chronic pain are maintained or enhanced by activity in the sympathetic nervous system (SNS), but attempts to model this have yielded conflicting findings. The SNS has both pro- and anti-inflammatory effects on immunity, confounding the interpretation of experiments using global sympathectomy methods. We performed a "microsympathectomy" by cutting the ipsilateral gray rami where they entered the spinal nerves near the L4 and L5 DRG. This led to profound sustained reductions in pain behaviors induced by local DRG inflammation (a rat model of low back pain) and by a peripheral paw inflammation model. Effects of microsympathectomy were evident within one day, making it unlikely that blocking sympathetic sprouting in the local DRGs or hindpaw was the sole mechanism. Prior microsympathectomy greatly reduced hyperexcitability of sensory neurons induced by local DRG inflammation observed 4 d later. Microsympathectomy reduced local inflammation and macrophage density in the affected tissues (as indicated by paw swelling and histochemical staining). Cytokine profiling in locally inflamed DRG showed increases in pro-inflammatory Type 1 cytokines and decreases in the Type 2 cytokines present at baseline, changes that were mitigated by microsympathectomy. Microsympathectomy was also effective in reducing established pain behaviors in the local DRG inflammation model. We conclude that the effect of sympathetic fibers in the L4/L5 gray rami in these models is pro-inflammatory. This raises the possibility that therapeutic interventions targeting gray rami might be useful in some chronic inflammatory pain conditions. SIGNIFICANCE STATEMENT Sympathetic blockade is used for many pain conditions, but preclinical studies show both pro- and anti-nociceptive effects. The sympathetic nervous system also has both pro- and anti-inflammatory effects on immune tissues and cells. We examined effects of a very localized sympathectomy. By cutting the gray rami to the spinal nerves near the lumbar sensory ganglia, we avoided widespread sympathetic denervation. This procedure profoundly reduced mechanical pain behaviors induced by a back pain model and a model of peripheral inflammatory pain. One possible mechanism was reduction of inflammation in the sympathetically denervated regions. This raises the possibility that therapeutic interventions targeting gray rami might be useful in some inflammatory conditions.
Collapse
|
30
|
Siebold C, Yamashita T, Monnier PP, Mueller BK, Pasterkamp RJ. RGMs: Structural Insights, Molecular Regulation, and Downstream Signaling. Trends Cell Biol 2017; 27:365-378. [PMID: 28007423 PMCID: PMC5404723 DOI: 10.1016/j.tcb.2016.11.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/27/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
Although originally discovered as neuronal growth cone-collapsing factors, repulsive guidance molecules (RGMs) are now known as key players in many fundamental processes, such as cell migration, differentiation, iron homeostasis, and apoptosis, during the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. Furthermore, three RGMs (RGMa, RGMb/DRAGON, and RGMc/hemojuvelin) have been linked to the pathogenesis of various disorders ranging from multiple sclerosis (MS) to cancer and juvenile hemochromatosis (JHH). While the molecular details of these (patho)biological effects and signaling modes have long remained unknown, recent studies unveil several exciting and novel aspects of RGM processing, ligand-receptor interactions, and downstream signaling. In this review, we highlight recent advances in the mechanisms-of-action and function of RGM proteins.
Collapse
Affiliation(s)
- Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Philippe P Monnier
- Krembil Research Institute, 60 Leonard Street, M5T 2S8, Toronto, ONT, Canada
| | - Bernhard K Mueller
- Neuroscience Discovery Research, Abbvie, Knollstrasse 50, 67061 Ludwigshafen, Germany
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Zhu Z, Otahal P, Wang B, Jin X, Laslett LL, Wluka AE, Antony B, Han W, Wang X, Winzenberg T, Cicuttini F, Jones G, Ding C. Cross-sectional and longitudinal associations between serum inflammatory cytokines and knee bone marrow lesions in patients with knee osteoarthritis. Osteoarthritis Cartilage 2017; 25:499-505. [PMID: 27836676 DOI: 10.1016/j.joca.2016.10.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 10/18/2016] [Accepted: 10/28/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To describe cross-sectional and longitudinal associations between serum levels of interleukin (IL) - 6, IL-17A, IL-17F, IL-23 and knee bone marrow lesions (BMLs) in patients with knee osteoarthritis (OA). DESIGN Patients (n = 192) with symptomatic knee OA (mean 63 years, range 50-79, female 53%) were assessed at baseline and after 24 months. At each time point, serum IL-6, IL-17A, IL-17F and IL-23 were measured using Bio-Plex® Multiplex Immunoassays with Luminex xMAP technology. Knee BMLs were scored using the modified whole organ MRI score (WORMS) from T2 weighted fat-suppressed fast spin echo magnetic resonance imaging (MRI). Multivariable linear regression and log binominal regression were used to determine the associations between cytokines and BMLs. RESULTS Baseline IL-6 (quarters) were significantly associated with total knee BMLs (P < 0.01 for the trend) as well as associated with an increase in BML score (P = 0.05 for the trend), after adjustment for confounders. Baseline IL-17F and IL-23 (highest quarters vs others) was associated with an increase in BML score in females (P = 0.04 for IL-17F; P = 0.01 for IL-23), but not in males, in multivariable analyses. In contrast, IL-17A was not significantly associated with BMLs in either females or males. CONCLUSION IL-6 is associated with increased knee BMLs in both females and males with OA. Serum IL-17F and IL-23 predicted increased knee BML scores in females only, suggesting that inflammation is involved in BML pathogenesis in knee OA, especially in women. TRAIL REGISTRATION ClinicalTrials.gov identifier: NCT01176344; Australian New Zealand Clinical Trials Registry: ACTRN12610000495022.
Collapse
Affiliation(s)
- Z Zhu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Arthritis Research Institute, 1(st) Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - P Otahal
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - B Wang
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - X Jin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - L L Laslett
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - A E Wluka
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - B Antony
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - W Han
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - X Wang
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Arthritis Research Institute, 1(st) Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - T Winzenberg
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - F Cicuttini
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - G Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - C Ding
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Arthritis Research Institute, 1(st) Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
32
|
Fujita Y, Yamashita T. The roles of RGMa-neogenin signaling in inflammation and angiogenesis. Inflamm Regen 2017; 37:6. [PMID: 29259705 PMCID: PMC5725648 DOI: 10.1186/s41232-017-0037-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/16/2017] [Indexed: 12/13/2022] Open
Abstract
Repulsive guidance molecule (RGM) is a glycosylphosphatidylinositol (GPI)-anchored glycoprotein that has diverse functions in the developing and pathological central nervous system (CNS). The binding of RGM to its receptor neogenin regulates axon guidance, neuronal differentiation, and survival during the development of the CNS. In the pathological state, RGM expression is induced after spinal cord injury, and the inhibition of RGM promotes axon growth and functional recovery. Furthermore, RGM expression is also observed in immune cells, and RGM regulates inflammation and neurodegeneration in autoimmune encephalomyelitis. RGMa induces T cell activation in experimental autoimmune encephalomyelitis (EAE), which is the animal model of multiple sclerosis (MS). RGM is expressed in pathogenic Th17 cells and induces neurodegeneration by binding to neogenin. Angiogenesis is an additional key factor involved in the pathophysiology of EAE. Via neogenin, treatment with RGMa can suppress endothelial tube formation; this finding indicates that RGMa inhibits neovascularization. These observations suggest the feasibility of utilizing the RGMa-neogenin signaling pathway as a therapeutic target to overcome inflammation and neurodegeneration. This review focuses on the molecular mechanisms of inflammation and angiogenesis via RGM-neogenin signaling.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
33
|
Menzel L, Paterka M, Bittner S, White R, Bobkiewicz W, van Horssen J, Schachner M, Witsch E, Kuhlmann T, Zipp F, Schäfer MKE. Down-regulation of neuronal L1 cell adhesion molecule expression alleviates inflammatory neuronal injury. Acta Neuropathol 2016; 132:703-720. [PMID: 27544757 DOI: 10.1007/s00401-016-1607-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/22/2016] [Accepted: 08/09/2016] [Indexed: 02/05/2023]
Abstract
In multiple sclerosis (MS), the immune cell attack leads to axonal injury as a major cause for neurological disability. Here, we report a novel role of the cell adhesion molecule L1 in the crosstalk between the immune and nervous systems. L1 was found to be expressed by CNS axons of MS patients and human T cells. In MOG35-55-induced murine experimental neuroinflammation, CD4+ T cells were associated with degenerating axons in the spinal cord, both expressing L1. However, neuronal L1 expression in the spinal cord was reduced, while levels of the transcriptional repressor REST (RE1-Silencing Transcription Factor) were up-regulated. In PLP139-151-induced relapsing-remitting neuroinflammation, L1 expression was low at the peak stage of disease, reached almost normal levels in the remission stage, but decreased again during disease relapse indicating adaptive expression regulation of L1. In vitro, activated CD4+ T cells caused contact-dependent down-regulation of L1, up-regulation of its repressor REST and axonal injury in co-cultured neurons. T cell adhesion to neurons and axonal injury were prevented by an antibody blocking L1 suggesting that down-regulation of L1 ameliorates neuroinflammation. In support of this hypothesis, antibody-mediated blocking of L1 in C57BL/6 mice as well as neuron-specific depletion of L1 in synapsinCre × L1fl/fl mice reduces disease severity and axonal pathology despite unchanged immune cell infiltration of the CNS. Our data suggest that down-regulation of neuronal L1 expression is an adaptive process of neuronal self-defense in response to pro-inflammatory T cells, thereby alleviating immune-mediated axonal injury.
Collapse
Affiliation(s)
- Lutz Menzel
- Department of Anesthesiology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Magdalena Paterka
- Department of Neurology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience (FTN) and Rhine Main Neuroscience Network (rmn²), Mainz, Germany
| | - Robin White
- Institute of Physiology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Wiesia Bobkiewicz
- Department of Anesthesiology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
| | - Esther Witsch
- Department of Neurology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Frauke Zipp
- Department of Neurology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neuroscience (FTN) and Rhine Main Neuroscience Network (rmn²), Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes-Gutenberg-University Mainz, Mainz, Germany.
- Focus Program Translational Neuroscience (FTN) and Rhine Main Neuroscience Network (rmn²), Mainz, Germany.
| |
Collapse
|
34
|
Xu X, Gao Y, Zhai Z, Zhang S, Shan F, Feng J. Repulsive guidance molecule a blockade exerts the immunoregulatory function in DCs stimulated with ABP and LPS. Hum Vaccin Immunother 2016; 12:2169-2180. [PMID: 26986456 PMCID: PMC4994741 DOI: 10.1080/21645515.2016.1164361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/24/2016] [Accepted: 03/05/2016] [Indexed: 02/01/2023] Open
Abstract
Repulsive guidance molecule a (RGMa) is an axonal guidance molecule that has recently found to exert function in immune system. This study evaluated the function of RGMa in modulation of dendritic cells (DCs) function stimulated with Achyranthes bidentata polysaccharide (ABP) and lipopolysaccharide (LPS) using a RGMa-neutralizing antibody. Compared with the Control-IgG/ABP and Control-IgG/LPS groups, DCs in the Anti-RGMa/ABP and Anti-RGMa/LPS groups 1) showed small, round cells with a few cell processes and organelles, and many pinocytotic vesicles; 2) had decreased MHC II, CD86, CD80, and CD40 expression; 3) displayed the decreased IL-12p70, IL-1β and TNF-α levels and increased IL-10 secretion; 4) had a high percentage of FITC-dextran uptake; and 5) displayed a reduced ability to drive T cell proliferation and reinforced T cell polarization toward a Th2 cytokine pattern. We conclude that DCs treated with RGMa-neutralizing antibodies present with tolerogenic and immunoregulatory characteristics, which provides new insights into further understanding of the function of RGMa.
Collapse
Affiliation(s)
- Xuxu Xu
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Yan Gao
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Zhiyong Zhai
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Shuo Zhang
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
35
|
Levin MC, Lee S, Gardner LA, Shin Y, Douglas JN, Salapa H. Autoantibodies to heterogeneous nuclear ribonuclear protein A1 (hnRNPA1) cause altered 'ribostasis' and neurodegeneration; the legacy of HAM/TSP as a model of progressive multiple sclerosis. J Neuroimmunol 2016; 304:56-62. [PMID: 27449854 DOI: 10.1016/j.jneuroim.2016.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/06/2016] [Indexed: 12/23/2022]
Abstract
Several years following its discovery in 1980, infection with human T-lymphotropic virus type 1 (HTLV-1) was shown to cause HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP), a disease biologically similar to progressive forms of multiple sclerosis (MS). In this manuscript, we review some of the clinical, pathological, and immunological similarities between HAM/TSP and MS with an emphasis on how autoantibodies to an RNA binding protein, heterogeneous nuclear ribonuclear protein A1 (hnRNP A1), might contribute to neurodegeneration in immune mediated diseases of the central nervous system.
Collapse
Affiliation(s)
- Michael C Levin
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Sangmin Lee
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lidia A Gardner
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yoojin Shin
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joshua N Douglas
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hannah Salapa
- Veterans Administration Medical Center, Memphis, TN, USA; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Anatomy/Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
36
|
Xu X, Gao Y, Shan F, Feng J. A novel role for RGMa in modulation of bone marrow-derived dendritic cells maturation induced by lipopolysaccharide. Int Immunopharmacol 2016; 33:99-107. [PMID: 26896667 DOI: 10.1016/j.intimp.2016.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/16/2016] [Accepted: 02/08/2016] [Indexed: 02/06/2023]
Abstract
Repulsive guidance molecule a (RGMa) is known to mediate immune responses and has been indicated to modulates T cell activation and autoimmune diseases by dendritic cells (DCs), which hints its significant function in the latter cells. The aim of our study, therefore, was to evaluate the function of RGMa in DC maturation. We found that small interfering RNA (siRNA) successfully silenced the expression of RGMa in DCs. Even after LPS stimulation, RGMa-silenced DCs displayed an immature morphology, characterized by small, round cells with a few cell processes and organelles, and many pinocytotic vesicles. In the presence of LPS, RGMa siRNA transfection markedly reduced levels of CD80, CD86, CD40, and MHC II expression, as well as the secretion of IL-12p70 and TNF-α. With LPS treatment, RGMa siRNA-transfected DCs also showed increased levels of IL-10 and endocytosis. Moreover, in the presence of LPS, RGMa siRNA-transfected DCs displayed a low ability to induce T cell proliferation and differentiation, compared with negative control (NTi)-transfected or control DCs (p<0.05 for both). We conclude that after LPS stimulation, RGMa siRNA-transfected DCs show immunoregulatory and tolerogenic characteristics, which provides new insights into the immune system.
Collapse
Affiliation(s)
- Xuxu Xu
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, PR China
| | - Yan Gao
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, PR China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang 110000, PR China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
37
|
Liu Y, Peng H, Meng Z, Wei M. Correlation of IL-17 Level in Synovia and Severity of Knee Osteoarthritis. Med Sci Monit 2015; 21:1732-6. [PMID: 26076201 PMCID: PMC4480114 DOI: 10.12659/msm.893771] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background The aim of this study was to evaluate the role of interleukin-17 (IL-17) level in synovia and its relationship with the severity of knee osteoarthritis (OA). Material/Methods We enrolled 226 OA patients and 106 controls in this study. The symptomatic/radiation severity of OA was assessed by the Western Ontario McMaster University Osteoarthritis Index (WOMAC) pain score/Kellgren-Lawrence (KL) grading system. Serum IL-17 levels were measured by enzyme-linked immunosorbent assay (ELISA). Results Synovia IL-17 levels were significantly higher in OA patients compared with controls (P<0.01), and were negatively correlated with OA severity. IL-17 level gradually decreased among different phases but lacked statistical significance. Conclusions IL-17 might play a crucial role in the pathogenesis of OA and is closely related to pain. Blocking the IL-17 signaling pathway may delay pain related to OA.
Collapse
Affiliation(s)
- Yingsong Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Hao Peng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Zhao Meng
- Department of Orthopedics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China (mainland)
| | - Mingzhu Wei
- Department of Orthopedics, Shiyan Hospital of Integrated Traditional and Western Medicine, Shiyan, Hubei, China (mainland)
| |
Collapse
|