1
|
Duncan ID, Vivian JA, August BK, Keuler NS, Komro A, Radecki D, Kiland JA, Gandhi R, Reilly M, Cameron S, Rylander H, Pritchard J, Ver Hoeve JN. Promotion of remyelination by a thyromimetic drug leading to functional recovery. Exp Neurol 2025; 389:115227. [PMID: 40120662 DOI: 10.1016/j.expneurol.2025.115227] [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: 03/06/2025] [Revised: 03/18/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Promotion of remyelination has become a critical therapeutic approach in the treatment of demyelinating disorders including multiple sclerosis (MS), both to restore function and protect intact axons against future degeneration. Thyroid hormone receptor agonist mediated signaling is critical for the maturation of oligodendrocytes (Ols) from oligodendrocyte precursor cells (OPCs) and may be a rational target for drug development in the treatment of MS. Therefore, we tested the potential of a thyromimetic drug pro-drug, LL-341070, to promote remyelination and neurologic recovery in a unique large animal model in which there is extensive demyelination throughout the CNS that results from the prolonged feeding of irradiated food. In four out of eight cats fed the irradiated diet that had developed significant neurologic dysfunction, daily treatment with LL-341070 led to clinical improvement or complete recovery of baseline function. Extensive evidence of remyelination was observed throughout the brain, spinal cord and in the optic nerve in these four animals when compared with non- treated animals. These results provide support for thyroid hormone receptor agonism as a potential novel target to promote remyelination and clinical outcomes in patients with MS.
Collapse
Affiliation(s)
- Ian D Duncan
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Jeffrey A Vivian
- Autobahn Therapeutics Inc, 9880 Campus Point Drive, San Diego, CA, United States of America
| | - Benjamin K August
- Electron Microscopy Facility, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Nicholas S Keuler
- Department of Statistics, College of Letters and Science, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Abigail Komro
- Electron Microscopy Facility, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Daniel Radecki
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Julie A Kiland
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Rohan Gandhi
- Autobahn Therapeutics Inc, 9880 Campus Point Drive, San Diego, CA, United States of America
| | - Madelyn Reilly
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Starr Cameron
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Helena Rylander
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - Jessica Pritchard
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States of America
| | - James N Ver Hoeve
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, United States of America.
| |
Collapse
|
2
|
Makhzoum H, El Majzoub R, Ismail A, Kassem M, Kotaich J, Chahine B. Interventions promoting remyelination in multiple sclerosis: a systematic review of clinical trials. Neurol Res 2025:1-18. [PMID: 40393003 DOI: 10.1080/01616412.2025.2507756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/10/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVES Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by demyelination and axonal damage. Current therapies primarily manage symptoms and slow disease progression but do not achieve remyelination. This systematic review evaluates the efficacy and safety of remyelination-promoting interventions in MS, focusing on clinical trials utilizing outcome measures validated in prior studies as indicators of remyelination. METHODS A comprehensive search of PubMed, Embase, Web of Science, and Cochrane Library was conducted in May 2024. Clinical trials assessing interventions with potential remyelinating properties were included. Data extraction followed standardized forms, and study quality was evaluated using ROBINS-I for non-randomized trials and RoB 2.0 for RCTs. Remyelination was assessed using Magnetization Transfer Ratio (MTR), Visual Evoked Potentials (VEPs), Myelin Water Fraction (MWF), Diffusion Tensor Imaging (DTI), and Optical Coherence Tomography (OCT). RESULTS From 1,615 screened records, 25 studies met the inclusion criteria and were analyzed. Across the 3341 participants, 17 interventions were evaluated. Most studies demonstrated a moderate risk of bias, yet all interventions, except one, were generally safe and well tolerated. Notably, rHIgM22, L-T3, opicinumab, clemastine fumarate, phenonytoin, domperidone, GSK239512, human fetal neural precursor cells (hfNPCs), and low-intensity repetitive transcranial magnetic stimulation (LI-rTMS) exhibited remyelination potential. Additionally, disease-modifying therapies (DMTs) such as Ocrelizumab, Fingolimod, and Natalizumab showed promising effects. DISCUSSION Although several interventions demonstrated remyelination potential, limitations such as small sample sizes, short follow-up periods, and lack of standardized clinical endpoints validating remyelination's functional impact, highlight the need for robust clinical trial designs, advanced biomarkers, and combination therapies integrating remyelination, neuroprotection, and immunomodulation to improve MS treatment outcomes.
Collapse
Affiliation(s)
- Houssein Makhzoum
- Neuroscience Research Center, Faculty of Medical sciences, Lebanese University, Hadath, Lebanon
| | - Rania El Majzoub
- Neuroscience Research Center, Faculty of Medical sciences, Lebanese University, Hadath, Lebanon
- School of Pharmacy (Department of Biomedical Sciences), Lebanese International University, Mazraa, Lebanon
| | - Ali Ismail
- Neuroscience Research Center, Faculty of Medical sciences, Lebanese University, Hadath, Lebanon
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- Department of Neurology, MEDICA Research Investigation, Hadath, Lebanon
| | - Mariam Kassem
- Faculty of Arts and Sciences, Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Jana Kotaich
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- Department of Neurology, MEDICA Research Investigation, Hadath, Lebanon
| | - Bahia Chahine
- School of Pharmacy (Department of Biomedical Sciences), Lebanese International University, Mazraa, Lebanon
| |
Collapse
|
3
|
Gluck L, Gerstein B, Kaunzner UW. Repair mechanisms of the central nervous system: From axon sprouting to remyelination. Neurotherapeutics 2025:e00583. [PMID: 40348704 DOI: 10.1016/j.neurot.2025.e00583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
The central nervous system (CNS), comprising the brain, spinal cord, and optic nerve, has limited regenerative capacity, posing significant challenges in treating neurological disorders. Recent advances in neuroscience and neurotherapeutics have introduced promising strategies to stimulate CNS repair, particularly in the context of neurodegenerative diseases such as multiple sclerosis. This review explores the complex interplay between inflammation, demyelination, and remyelination possibilities. Glial cells, including oligodendrocyte precursors, oligodendrocytes, astrocytes and microglia play dual roles in injury response, with reactive gliosis promoting repair but also potentially inhibiting recovery through glial scar formation. There is also an emphasis on axonal regeneration, axonal sprouting and stem cell therapies. We highlight the role of neuroplasticity in recovery post-injury and the limited regenerative potential of axons in the CNS due to inhibitory factors such as myelin-associated inhibitors. Moreover, neurotrophic factors support neuronal survival and axonal growth, while stem cell-based approaches offer promise for replacing lost neurons and glial cells. However, challenges such as stem cell survival, integration, and risk of tumor formation remain. Furthermore, we examine the role of neurogenesis in CNS repair and the remodeling of the extracellular matrix, which can facilitate regeneration. Through these diverse mechanisms, ongoing research aims to overcome the intrinsic and extrinsic barriers to CNS repair and advance therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lauren Gluck
- Montefiore Medical Center, 1250 Waters Place Tower 2, Bronx, NY 10461, USA.
| | - Brittany Gerstein
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| | - Ulrike W Kaunzner
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| |
Collapse
|
4
|
Albelo-Martínez M, Rizvi S. Progressive multiple sclerosis: Evaluating current therapies and exploring future treatment strategies. Neurotherapeutics 2025:e00601. [PMID: 40345951 DOI: 10.1016/j.neurot.2025.e00601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/13/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Progressive forms of multiple sclerosis (MS) include primary progressive MS (PPMS) and secondary progressive MS (SPMS). Unlike relapsing-remitting MS (RRMS), progressive MS is recognized by relentless progression with accumulating disability, rare to no relapses nor new activity on MRIs. Clinically, neurologic worsening in MS can occur in the relapsing-remitting (RRMS) phase of disease due to incomplete recovery from neuroinflammatory relapses. However, a progressive disease course is the dominant factor related to accumulating disability. There is persistent central nervous system (CNS) compartmentalized inflammation, mitochondrial dysfunction and altered immune responses. Unlike in RRMS, the efficacy of disease modifying agents (DMA) in progressive MS has been limited, highlighting the need for novel therapeutic approaches that address both inflammation and neurodegeneration. This article explores current management of progressive MS, and future directions in targeting the unique pathophysiology of this complex disease.
Collapse
Affiliation(s)
- Marelisa Albelo-Martínez
- Department of Neurology, Brown University Health and Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| | - Syed Rizvi
- Department of Neurology, Brown University Health and Rhode Island Hospital, Alpert Medical School of Brown University, USA.
| |
Collapse
|
5
|
Liu K, Chen X, Fan Z, Ren F, Liu J, Hu B. From organoids to organoids-on-a-chip: Current applications and challenges in biomedical research. Chin Med J (Engl) 2025; 138:792-807. [PMID: 39994843 PMCID: PMC11970821 DOI: 10.1097/cm9.0000000000003535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Indexed: 02/26/2025] Open
Abstract
ABSTRACT The high failure rates in clinical drug development based on animal models highlight the urgent need for more representative human models in biomedical research. In response to this demand, organoids and organ chips were integrated for greater physiological relevance and dynamic, controlled experimental conditions. This innovative platform-the organoids-on-a-chip technology-shows great promise in disease modeling, drug discovery, and personalized medicine, attracting interest from researchers, clinicians, regulatory authorities, and industry stakeholders. This review traces the evolution from organoids to organoids-on-a-chip, driven by the necessity for advanced biological models. We summarize the applications of organoids-on-a-chip in simulating physiological and pathological phenotypes and therapeutic evaluation of this technology. This section highlights how integrating technologies from organ chips, such as microfluidic systems, mechanical stimulation, and sensor integration, optimizes organoid cell types, spatial structure, and physiological functions, thereby expanding their biomedical applications. We conclude by addressing the current challenges in the development of organoids-on-a-chip and offering insights into the prospects. The advancement of organoids-on-a-chip is poised to enhance fidelity, standardization, and scalability. Furthermore, the integration of cutting-edge technologies and interdisciplinary collaborations will be crucial for the progression of organoids-on-a-chip technology.
Collapse
Affiliation(s)
- Kailun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaowei Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Fan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Ren
- State Key Lab of Processors, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101 China
| |
Collapse
|
6
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
7
|
Alsubhi AH, Althagafi MK, Alhamed L, Alothman RA, Alothman AAA, Alghorair AS, Alruwaili SA, Badeeb N. Exploring the efficacy and safety of neuroprotective agents in optic neuritis: a systematic review and meta-analysis. Int Ophthalmol 2025; 45:80. [PMID: 39969696 DOI: 10.1007/s10792-025-03447-z] [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: 07/26/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE This review aims to systematically review the available evidence on neuroprotective agents for optic neuritis, evaluating their effectiveness and safety. METHODS This systematic review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guidelines. To collect the studies, we searched electronic databases including MEDLINE, Cochrane Library, and EMBASE in May 2023. A pre-established protocol of the systemic review was registered in the International Prospective Register of Systematic Reviews, ID Number: (CRD42023448479). RESULTS Regarding the global loss of RNFL, a statistical significance was observed in favor of erythropoietin compared to placebo (MD = - 5.40, 95%CI, - 7.27-- 3.53). The overall difference in VEP latency recovery was insignificant (MD = - 0.63, 95%CI, - 5.95-4.69). The distinctness in visual acuity measured in logMAR in the memantine group was significant (MD = - 0.25, 95%CI, - 0.33-- 0.16). Two of the included studies had no major adverse events, however, the likelihood of major adverse events was significant overall in the other studies (RR = 2.28, 95%CI, 1.16 to 4.47). The erythropoietin group had 12 cases of major adverse events with a RR = 2.35. CONCLUSION Evaluating neuroprotective agents for ON produced mixed findings. Although erythropoietin was effective in reducing the thinning of RNFL, it did not lead to improved visual acuity results. On the other hand, our analysis found that memantine significantly improved VA compared to placebo. Notably, the experimental groups had more frequent adverse events, particularly with erythropoietin. Thus, caution is advised when contemplating the use of these agents.
Collapse
Affiliation(s)
| | | | - Latifah Alhamed
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | | | | | - Nooran Badeeb
- Department of Surgery, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Ricigliano VAG, Marenna S, Borrelli S, Camera V, Carnero Contentti E, Szejko N, Bakirtzis C, Gluscevic S, Samadzadeh S, Hartung HP, Selmaj K, Stankoff B, Comi G, ECF Young Investigators/Fellows Initiative. Identifying Biomarkers for Remyelination and Recovery in Multiple Sclerosis: A Measure of Progress. Biomedicines 2025; 13:357. [PMID: 40002770 PMCID: PMC11853245 DOI: 10.3390/biomedicines13020357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Multiple sclerosis (MS) pathology is characterized by acute and chronic inflammation, demyelination, axonal injury, and neurodegeneration. After decades of research into MS-related degeneration, recent efforts have shifted toward recovery and the prevention of further damage. A key area of focus is the remyelination process, where researchers are studying the effects of pharmacotherapy on myelin repair mechanisms. Multiple compounds are being tested for their potential to foster remyelination in different clinical settings through the application of less or more complex techniques to assess their efficacy. Objective: To review current methods and biomarkers to track myelin regeneration and recovery over time in people with MS (PwMS), with potential implications for promyelinating drug testing. Methods: Narrative review, based on a selection of PubMed articles discussing techniques to measure in vivo myelin repair and functional recovery in PwMS. Results: Non-invasive tools, such as structural Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET), are being implemented to track myelin repair, while other techniques like evoked potentials, functional MRI, and digital markers allow the assessment of functional recovery. These methods, alone or in combination, have been employed to obtain precise biomarkers of remyelination and recovery in various clinical trials on MS. Conclusions: Combining different techniques to identify myelin restoration in MS could yield novel biomarkers, enhancing the accuracy of clinical trial outcomes for remyelinating therapies in PwMS.
Collapse
Affiliation(s)
- Vito A. G. Ricigliano
- Neurology Department, Pitié-Salpêtrière Hospital, APHP, 75013 Paris, France; (V.A.G.R.); (B.S.)
- Neurology Unit, GHNE—Paris Saclay Hospital, 91400 Orsay, France
| | - Silvia Marenna
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), IRCCS-San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, 20132 Milan, Italy;
| | - Serena Borrelli
- Neuroinflammation Imaging Laboratory (NIL), Institute of NeuroScience, Université Catholique de Louvain, 1348 Brussels, Belgium;
- Department of Neurology, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Valentina Camera
- Nuffield Departement of Clinical Neuroscience, University of Oxford, Oxford OX1 2JD, UK;
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, 37129 Verona, Italy
| | - Edgar Carnero Contentti
- Neuroimmunology Unit, Department of Neurosciences, Hospital Alemán, Buenos Aires C1425FQB, Argentina;
| | - Natalia Szejko
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Bioethics, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Christos Bakirtzis
- Multiple Sclerosis Center, Second Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Sanja Gluscevic
- Neurology Clinic, Clinical Centre of Montenegro, 81000 Podgorica, Montenegro;
| | - Sara Samadzadeh
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Institute of Regional Health Research and, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- The Center for Neurological Research, Department of Neurology, Næstved-Slagelse-Ringsted Hospitals, 4200 Slagelse, Denmark
| | - Hans-Peter Hartung
- Brain and Mind Center, University of Sydney, Sydney 2050, Australia;
- Department of Neurology, Palacky University Olomouc, 779 00 Olomouc, Czech Republic
- Department of Neurology, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Krzysztof Selmaj
- Department of Neurology, University of Warmia & Mazury, 10-719 Olsztyn, Poland;
| | - Bruno Stankoff
- Neurology Department, Pitié-Salpêtrière Hospital, APHP, 75013 Paris, France; (V.A.G.R.); (B.S.)
- Paris Brain Institute, ICM, CNRS, Inserm, Sorbonne Université, 75005 Paris, France
| | - Giancarlo Comi
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, 20129 Milan, Italy;
| | | |
Collapse
|
9
|
Gokoffski KK, Washington KM, Chuck RS. Clinical and Scientific Considerations for Whole Eye Transplantation: An Ophthalmologist's Perspective. Transl Vis Sci Technol 2025; 14:13. [PMID: 39918461 PMCID: PMC11809445 DOI: 10.1167/tvst.14.2.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Affiliation(s)
- Kimberly K Gokoffski
- Department of Ophthalmology, University of Southern California, Los Angeles, CA, USA
| | - Kia M Washington
- Division of Plastic and Reconstructive Surgery, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - Roy S Chuck
- Department of Ophthalmology, Albert Einstein College of Medicine, Bronx, NY, USA
- Editor-in-Chief
| |
Collapse
|
10
|
Tong B, Zhang X, Hu H, Yang H, Wang X, Zhong M, Yang F, Hua F. From diagnosis to treatment: exploring the mechanisms underlying optic neuritis in multiple sclerosis. J Transl Med 2025; 23:87. [PMID: 39838397 PMCID: PMC11748848 DOI: 10.1186/s12967-025-06105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease affecting the central nervous system, commonly causing sensory disturbances, motor weakness, impaired gait, incoordination and optic neuritis (ON). According to the statistics, up to 50% of MS patients experience vision problems during the disease course, suffering from blurred vision, pain, color vision deficits, and even blindness. Treatments have progressed from corticosteroids to therapies targeted against B/T cells. This review comprehensively and systematically reappraises the diagnostic methods for visual impairment in MS patients. It also summarizes the most recent treatment approaches and effective medications for ON in MS. Finally, we examine the immunoinflammatory mechanisms that underlie lesions in the central nervous system in multiple sclerosis, in order to direct future investigations to confirm these mechanisms in the visual pathway.
Collapse
Affiliation(s)
- Bin Tong
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 34100, Jiangxi, People's Republic of China
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Xin Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 34100, Jiangxi, People's Republic of China
| | - Haijian Hu
- Department of Ophthalmology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Huayi Yang
- Nanchang Medical College, Nanchang, 330004, Jiangxi, People's Republic of China
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Maolin Zhong
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 34100, Jiangxi, People's Republic of China
| | - Fan Yang
- Department of Cardiothoracic Surgery, People's Hospital of Ruijin City, Ruijin, 342500, Jiangxi, People's Republic of China.
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 34100, Jiangxi, People's Republic of China.
- Jiangxi Provincial Key Laboratory of Anesthesiology, 1# Minde Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
11
|
Strecker J, Schmidt‐Pogoda A, Diederich K, Zaremba D, Wieters F, Beuker C, Koecke MHM, Straeten FA, Wiendl H, Minnerup J. Anti-LINGO-1 treatment restores myelination of corticospinal tract neurons and improves functional recovery after stroke. Brain Pathol 2025; 35:e13280. [PMID: 38946137 PMCID: PMC11669405 DOI: 10.1111/bpa.13280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Demyelination of corticospinal tract neurons contributes to long-term disability after cortical stroke. Nonetheless, poststroke myelin loss has not been addressed as a therapeutic target, so far. We hypothesized that an antibody-mediated inhibition of the Nogo receptor-interacting protein (LINGO-1, leucine-rich repeat and immunoglobulin domain-containing Nogo receptor-interacting protein) may counteract myelin loss, enhance remyelination and axonal growth, and thus promote functional recovery following stroke. To verify this hypothesis, mice were subjected to photothrombotic stroke and received either an antibody against LINGO-1 (n = 19) or a control treatment (n = 18). Behavioral tests were performed to assess the effects of anti-LINGO-1 treatment on the functional recovery. Seven weeks after stroke, immunohistochemical analyses were performed to analyze the effect of anti-LINGO-1 treatment on myelination and axonal loss of corticospinal tract neurons, proliferation of oligodendrocytes and neurogenesis. Anti-LINGO-1 treatment resulted in significantly improved functional recovery (p < 0.0001, repeated measures analysis of variance), and increased neurogenesis in the hippocampus and subventricular zone of the ipsilateral hemisphere (p = 0.0094 and p = 0.032, t-test). Notably, we observed a significant increase in myelin (p = 0.0295, t-test), platelet-derived growth factor receptor α-positive oligodendrocyte precursor cells (p = 0.0356, t-test) and myelinating adenomatous polyposis coli-positive cells within the ipsilateral internal capsule of anti-LINGO-1-treated mice (p = 0.0021, t-test). In conclusion, we identified anti-LINGO-1 as the first neuroregenerative treatment that counteracts poststroke demyelination of corticospinal tract neurons, presumably by increased proliferation of myelin precursor cells, and thereby improves functional recovery. Most importantly, our study presents myelin loss as a novel therapeutic target following stroke.
Collapse
Affiliation(s)
- Jan‐Kolja Strecker
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Antje Schmidt‐Pogoda
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Kai Diederich
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Dario Zaremba
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Frederique Wieters
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Carolin Beuker
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | | | - Frederike Anne Straeten
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Heinz Wiendl
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| | - Jens Minnerup
- Department of Neurology, Institute of Translational NeurologyUniversity of MünsterMünsterGermany
| |
Collapse
|
12
|
Aktas O, Ziemssen F, Ziemssen T, Klistorner A, Butzkueven H, Izquierdo G, Leocani L, Balcer LJ, Galetta SL, Castrillo-Viguera C, Bradley DP, Naylor ML, Belachew S, Franchimont N, Zhu B, Cheng W. RENEWED: A follow-up study of the opicinumab phase 2 RENEW study in participants with acute optic neuritis. Mult Scler Relat Disord 2025; 93:106185. [PMID: 39662163 DOI: 10.1016/j.msard.2024.106185] [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: 03/01/2024] [Revised: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND The randomized, phase 2 RENEW trial (NCT01721161) evaluated efficacy/safety of opicinumab (anti-LINGO-1) versus placebo in patients with first-episode unilateral acute optic neuritis (AON). Although no significant differences in the latency recovery of visual evoked potential (VEP) were observed between opicinumab and placebo groups in the intention to treat (ITT) population, the prespecified per-protocol (PP) population showed better recovery with opicinumab than with placebo. RENEWED (NCT02657915) was a one-visit, follow-up study 2 years after the last RENEW study visit (Week 32) designed to assess the long-term electrophysiological and clinical outcomes for participants previously enrolled and having received study treatment in RENEW. METHODS In the original study (RENEW), participants (aged 18-55 years) with a first unilateral AON episode were enrolled ≤28 days from first symptom onset and after treatment with methylprednisolone 1 g/day intravenously for 3-5 days; these participants were randomized to receive opicinumab 100 mg/kg or placebo intravenously once every 4 weeks from baseline to Week 20, assessed up to Week 32. Participants who received ≥1 dose of opicinumab 100 mg/kg or placebo in RENEW were eligible for the RENEWED follow-up study. Participants enrolled in RENEWED at 2 years (with an additional up to 12-month window) after the last RENEW study visit (Week 32) in both ITT and PP populations. The primary endpoint was change in full-field VEP (FF-VEP) latency of the affected eye at RENEWED study visit versus baseline of the fellow eye in RENEW, comparing between participants who received opicinumab and placebo in RENEW. Clinical progression and severity of multiple sclerosis (MS) were assessed. A substudy evaluated latency recovery using multifocal VEP (mfVEP) as an exploratory endpoint. RESULTS Of 82 RENEW participants, 52 (63.4 %; opicinumab n = 28, placebo n = 24) enrolled in and completed RENEWED. The adjusted mean (95 % CI) difference in FF-VEP latency delay between opicinumab and placebo groups was -6.0 (-14.6, 2.6) msec (p = 0.165) for the PP population and -4.5 (-12.6, 3.7) msec (p = 0.274) for the ITT population at the RENEWED study visit. Nominally significant improvement on mfVEP latency in the opicinumab group versus placebo was observed in participants of the mfVEP substudy (p = 0.009). In participants from the PP population without clinically definite MS (CDMS) at RENEW baseline,12 (55 %) in the opicinumab group and 12 (67 %) in the placebo group developed CDMS from enrollment in the RENEW study up to RENEWED Day 1; the estimated proportion of participants with CDMS at 2 years after the last study visit assessment in RENEW was lower when treated with opicinumab (0.50) than when treated with placebo (0.61) (hazard ratio p-value = 0.23). No benefit on visual acuity or other neurological functions was observed in the opicinumab group vs placebo in RENEWED. CONCLUSION The numerically increased VEP latency recovery with opicinumab treatment in RENEWED was consistent with those observed in the parent study RENEW. However, the VEP latency and clinical data in RENEWED should be interpreted with caution, given the nature of the follow-up study, the small sample size and the limitation in study design.
Collapse
Affiliation(s)
- Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Focke Ziemssen
- Center for Ophthalmology, Eberhard Karls University of Tübingen, Tübingen, Germany; Department of Ophthalmology, University Hospital Leipzig, Leipzig, Germany
| | - Tjalf Ziemssen
- MS Center Dresden, Center of Clinical Neuroscience, Dresden University of Technology, Dresden, Germany
| | - Alexander Klistorner
- Clinical Medicine, Macquarie University, Sydney, NSW, Australia; Save Sight Institute, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University Alfred Campus, Melbourne, VIC, Australia
| | - Guillermo Izquierdo
- Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Letizia Leocani
- University Vita-Salute San Raffaele, Milan, Italy; IRCCS Scientific Institute San Raffaele, Milan, Italy; Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Laura J Balcer
- Departments of Neurology, Population Health and Ophthalmology, NYU School of Medicine, New York, NY, USA
| | - Steven L Galetta
- Departments of Neurology, Population Health and Ophthalmology, NYU School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Sharma T, Mehan S, Tiwari A, Khan Z, Gupta GD, Narula AS. Targeting Oligodendrocyte Dynamics and Remyelination: Emerging Therapies and Personalized Approaches in Multiple Sclerosis Management. Curr Neurovasc Res 2025; 21:359-417. [PMID: 39219420 DOI: 10.2174/0115672026336440240822063430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/01/1970] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a progressive autoimmune condition that primarily affects young people and is characterized by demyelination and neurodegeneration of the central nervous system (CNS). This in-depth review explores the complex involvement of oligodendrocytes, the primary myelin- producing cells in the CNS, in the pathophysiology of MS. It discusses the biochemical processes and signalling pathways required for oligodendrocytes to function and remain alive, as well as how they might fail and cause demyelination to occur. We investigate developing therapeutic options that target remyelination, a fundamental component of MS treatment. Remyelination approaches promote the survival and differentiation of oligodendrocyte precursor cells (OPCs), restoring myelin sheaths. This improves nerve fibre function and may prevent MS from worsening. We examine crucial parameters influencing remyelination success, such as OPC density, ageing, and signalling pathway regulation (e.g., Retinoid X receptor, LINGO-1, Notch). The review also examines existing neuroprotective and antiinflammatory medications being studied to see if they can assist oligodendrocytes in surviving and reducing the severity of MS symptoms. The review focuses on medicines that target the myelin metabolism in oligodendrocytes. Altering oligodendrocyte metabolism has been linked to reversing demyelination and improving MS patient outcomes through various mechanisms. We also explore potential breakthroughs, including innovative antisense technologies, deep brain stimulation, and the impact of gut health and exercise on MS development. The article discusses the possibility of personalized medicine in MS therapy, emphasizing the importance of specific medicines based on individual molecular profiles. The study emphasizes the need for reliable biomarkers and improved imaging tools for monitoring disease progression and therapy response. Finally, this review focuses on the importance of oligodendrocytes in MS and the potential for remyelination therapy. It also underlines the importance of continued research to develop more effective treatment regimens, taking into account the complexities of MS pathology and the different factors that influence disease progression and treatment.
Collapse
Affiliation(s)
- Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
14
|
Garton T, Gadani SP, Gill AJ, Calabresi PA. Neurodegeneration and demyelination in multiple sclerosis. Neuron 2024; 112:3231-3251. [PMID: 38889714 PMCID: PMC11466705 DOI: 10.1016/j.neuron.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Progressive multiple sclerosis (PMS) is an immune-initiated neurodegenerative condition that lacks effective therapies. Although peripheral immune infiltration is a hallmark of relapsing-remitting MS (RRMS), PMS is associated with chronic, tissue-restricted inflammation and disease-associated reactive glial states. The effector functions of disease-associated microglia, astrocytes, and oligodendrocyte lineage cells are beginning to be defined, and recent studies have made significant progress in uncovering their pathologic implications. In this review, we discuss the immune-glia interactions that underlie demyelination, failed remyelination, and neurodegeneration with a focus on PMS. We highlight the common and divergent immune mechanisms by which glial cells acquire disease-associated phenotypes. Finally, we discuss recent advances that have revealed promising novel therapeutic targets for the treatment of PMS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Thomas Garton
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sachin P Gadani
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander J Gill
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Calabresi
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
McMurran CE, Mukherjee T, Brown JWL, Coles AJ, Cunniffe NG. Bexarotene leads to durable improvements in visual evoked potential latency: A follow-up study of the Cambridge Centre for Myelin Repair One trial. Mult Scler 2024; 30:1066-1071. [PMID: 38426437 PMCID: PMC11290016 DOI: 10.1177/13524585241233177] [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: 11/29/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
The Cambridge Centre for Myelin Repair One (CCMR-One) trial showed that 6 months of bexarotene reduces visual evoked potential (VEP) latency in people with relapsing-remitting multiple sclerosis (MS). In a single-centre follow-up study of these participants, we re-examined full-field VEP and clinical assessments. Twenty participants (12 bexarotene and 8 placebo) were seen on average 27 months after their trial involvement. In an analysis of all eyes with recordable signal (24 bexarotene and 14 placebo), the adjusted bexarotene-placebo treatment difference in P100 latency was -7.79 (95% confidence interval (CI) = -14.76, -0.82) ms, p = 0.044. We conclude that there were durable improvements in VEP latency, suggesting long-term benefits from exposure to a remyelinating drug.
Collapse
Affiliation(s)
| | - Trisha Mukherjee
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - J William L Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge. UK/NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London. UK/Clinical Outcomes Research (CORe) Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Nick G Cunniffe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Bourdette D, Wooliscroft L. Developing drugs that promote remyelination: Is our in vitro screening approach too simplistic? Neurotherapeutics 2024; 21:e00386. [PMID: 38937160 PMCID: PMC11284535 DOI: 10.1016/j.neurot.2024.e00386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Affiliation(s)
- Dennis Bourdette
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Lindsey Wooliscroft
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; Neurology Service, Portland VA Health Care System, Portland, OR 97239, USA
| |
Collapse
|
17
|
Zuroff LR, Green AJ. The Study of Remyelinating Therapies in Multiple Sclerosis: Visual Outcomes as a Window Into Repair. J Neuroophthalmol 2024; 44:143-156. [PMID: 38654413 DOI: 10.1097/wno.0000000000002149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Amelioration of disability in multiple sclerosis requires the development of complementary therapies that target neurodegeneration and promote repair. Remyelination is a promising neuroprotective strategy that may protect axons from damage and subsequent neurodegeneration. METHODS A review of key literature plus additional targeted search of PubMed and Google Scholar was conducted. RESULTS There has been a rapid expansion of clinical trials studying putative remyelinating candidates, but further growth of the field is limited by the lack of consensus on key aspects of trial design. We have not yet defined the ideal study population, duration of therapy, or the appropriate outcome measures to detect remyelination in humans. The varied natural history of multiple sclerosis, coupled with the short time frame of phase II clinical trials, requires that we develop and validate biomarkers of remyelination that can serve as surrogate endpoints in clinical trials. CONCLUSIONS We propose that the visual system may be the most well-suited and validated model for the study potential remyelinating agents. In this review, we discuss the pathophysiology of demyelination and summarize the current clinical trial landscape of remyelinating agents. We present some of the challenges in the study of remyelinating agents and discuss current potential biomarkers of remyelination and repair, emphasizing both established and emerging visual outcome measures.
Collapse
Affiliation(s)
- Leah R Zuroff
- Department of Neurology (LZ), Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; and Department of Neurology (AJG), University of California San Francisco, San Francisco, California
| | | |
Collapse
|
18
|
Marziali LN, Hwang Y, Palmisano M, Cuenda A, Sim FJ, Gonzalez A, Volsko C, Dutta R, Trapp BD, Wrabetz L, Feltri ML. p38γ MAPK delays myelination and remyelination and is abundant in multiple sclerosis lesions. Brain 2024; 147:1871-1886. [PMID: 38128553 PMCID: PMC11068213 DOI: 10.1093/brain/awad421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 10/05/2023] [Accepted: 11/12/2023] [Indexed: 12/23/2023] Open
Abstract
Multiple sclerosis is a chronic inflammatory disease in which disability results from the disruption of myelin and axons. During the initial stages of the disease, injured myelin is replaced by mature myelinating oligodendrocytes that differentiate from oligodendrocyte precursor cells. However, myelin repair fails in secondary and chronic progressive stages of the disease and with ageing, as the environment becomes progressively more hostile. This may be attributable to inhibitory molecules in the multiple sclerosis environment including activation of the p38MAPK family of kinases. We explored oligodendrocyte precursor cell differentiation and myelin repair using animals with conditional ablation of p38MAPKγ from oligodendrocyte precursors. We found that p38γMAPK ablation accelerated oligodendrocyte precursor cell differentiation and myelination. This resulted in an increase in both the total number of oligodendrocytes and the migration of progenitors ex vivo and faster remyelination in the cuprizone model of demyelination/remyelination. Consistent with its role as an inhibitor of myelination, p38γMAPK was significantly downregulated as oligodendrocyte precursor cells matured into oligodendrocytes. Notably, p38γMAPK was enriched in multiple sclerosis lesions from patients. Oligodendrocyte progenitors expressed high levels of p38γMAPK in areas of failed remyelination but did not express detectable levels of p38γMAPK in areas where remyelination was apparent. Our data suggest that p38γ could be targeted to improve myelin repair in multiple sclerosis.
Collapse
Affiliation(s)
- Leandro N Marziali
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Yoonchan Hwang
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Marilena Palmisano
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ana Cuenda
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid 28049, Spain
| | - Fraser J Sim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Alberto Gonzalez
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Christina Volsko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Lawrence Wrabetz
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Maria L Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Università degli studi di Milano, Biometra department and IRCcs Carlo Besta, Milano 20133, Italy
| |
Collapse
|
19
|
Papadopoulou A, Pfister A, Tsagkas C, Gaetano L, Sellathurai S, D'Souza M, Cerdá-Fuertes N, Gugleta K, Descoteaux M, Chakravarty MM, Fuhr P, Kappos L, Granziera C, Magon S, Sprenger T, Hardmeier M. Visual evoked potentials in multiple sclerosis: P100 latency and visual pathway damage including the lateral geniculate nucleus. Clin Neurophysiol 2024; 161:122-132. [PMID: 38461596 DOI: 10.1016/j.clinph.2024.02.020] [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: 10/07/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE To explore associations of the main component (P100) of visual evoked potentials (VEP) to pre- and postchiasmatic damage in multiple sclerosis (MS). METHODS 31 patients (median EDSS: 2.5), 13 with previous optic neuritis (ON), and 31 healthy controls had VEP, optical coherence tomography and magnetic resonance imaging. We tested associations of P100-latency to the peripapillary retinal nerve fiber layer (pRNFL), ganglion cell/inner plexiform layers (GCIPL), lateral geniculate nucleus volume (LGN), white matter lesions of the optic radiations (OR-WML), fractional anisotropy of non-lesional optic radiations (NAOR-FA), and to the mean thickness of primary visual cortex (V1). Effect sizes are given as marginal R2 (mR2). RESULTS P100-latency, pRNFL, GCIPL and LGN in patients differed from controls. Within patients, P100-latency was significantly associated with GCIPL (mR2 = 0.26), and less strongly with OR-WML (mR2 = 0.17), NAOR-FA (mR2 = 0.13) and pRNFL (mR2 = 0.08). In multivariate analysis, GCIPL and NAOR-FA remained significantly associated with P100-latency (mR2 = 0.41). In ON-patients, P100-latency was significantly associated with LGN volume (mR2 = -0.56). CONCLUSIONS P100-latency is affected by anterior and posterior visual pathway damage. In ON-patients, damage at the synapse-level (LGN) may additionally contribute to latency delay. SIGNIFICANCE Our findings corroborate post-chiasmatic contributions to the VEP-signal, which may relate to distinct pathophysiological mechanisms in MS.
Collapse
Affiliation(s)
- Athina Papadopoulou
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Clinical Research, University of Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Armanda Pfister
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Charidimos Tsagkas
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Shaumiya Sellathurai
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Clinical Research, University of Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Marcus D'Souza
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland; Neurostatus AG, University Hospital of Basel, Basel, Switzerland
| | - Nuria Cerdá-Fuertes
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Clinical Research, University of Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland; Neurostatus AG, University Hospital of Basel, Basel, Switzerland
| | - Konstantin Gugleta
- University Eye Clinic Basel, University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Mallar M Chakravarty
- Douglas Mental Health University Institute, Departments of Psychiatry and Biomedical Engineering (M.M.C.), McGill University, Montreal, University of Sherbrooke (M.D.), Canada
| | - Peter Fuhr
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Clinical Research, University of Basel, Switzerland
| | - Ludwig Kappos
- Department of Clinical Research, University of Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Cristina Granziera
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland; Translational Imaging in Neurology (ThINK) Basel, Department of Biomedical Engineering, University of Basel, Basel, Switzerland; Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Stefano Magon
- Pharma Research and Early Development, Neuroscience and Rare Diseases Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Martin Hardmeier
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
20
|
Parrilla GE, Gupta V, Wall RV, Salkar A, Basavarajappa D, Mirzaei M, Chitranshi N, Graham SL, You Y. The role of myelin in neurodegeneration: implications for drug targets and neuroprotection strategies. Rev Neurosci 2024; 35:271-292. [PMID: 37983528 DOI: 10.1515/revneuro-2023-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023]
Abstract
Myelination of axons in the central nervous system offers numerous advantages, including decreased energy expenditure for signal transmission and enhanced signal speed. The myelin sheaths surrounding an axon consist of a multi-layered membrane that is formed by oligodendrocytes, while specific glycoproteins and lipids play various roles in this formation process. As beneficial as myelin can be, its dysregulation and degeneration can prove detrimental. Inflammation, oxidative stress, and changes in cellular metabolism and the extracellular matrix can lead to demyelination of these axons. These factors are hallmark characteristics of certain demyelinating diseases including multiple sclerosis. The effects of demyelination are also implicated in primary degeneration in diseases such as glaucoma and Alzheimer's disease, as well as in processes of secondary degeneration. This reveals a relationship between myelin and secondary processes of neurodegeneration, including resultant degeneration following traumatic injury and transsynaptic degeneration. The role of myelin in primary and secondary degeneration is also of interest in the exploration of strategies and targets for remyelination, including the use of anti-inflammatory molecules or nanoparticles to deliver drugs. Although the use of these methods in animal models of diseases have shown to be effective in promoting remyelination, very few clinical trials in patients have met primary end points. This may be due to shortcomings or considerations that are not met while designing a clinical trial that targets remyelination. Potential solutions include diversifying disease targets and requiring concomitant interventions to promote rehabilitation.
Collapse
Affiliation(s)
- Gabriella E Parrilla
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Vivek Gupta
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Roshana Vander Wall
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Akanksha Salkar
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Mehdi Mirzaei
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Nitin Chitranshi
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
| | - Stuart L Graham
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| | - Yuyi You
- Faculty of Human, Health, and Medical Science, Department of Clinical Medicine, Macquarie University, Wallumattagal Campus, Macquarie Park, NSW 2109, Australia
- Save Sight Institute, University of Sydney, 8 Macquarie St, Sydney, NSW 2000, Australia
| |
Collapse
|
21
|
Lereim RR, Nytrova P, Guldbrandsen A, Havrdova EK, Myhr KM, Barsnes H, Berven FS. Natalizumab promotes anti-inflammatory and repair effects in multiple sclerosis. PLoS One 2024; 19:e0300914. [PMID: 38527011 PMCID: PMC10962820 DOI: 10.1371/journal.pone.0300914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Multiple sclerosis is an inflammatory and degenerative disease of the central nervous system leading to demyelination and axonal loss. Relapsing-remitting multiple sclerosis (RRMS) is commonly treated by anti-inflammatory drugs, where one of the most effective drugs to date is the monoclonal antibody natalizumab. METHODS The cerebrospinal fluid (CSF) proteome was analyzed in 56 patients with RRMS before and after natalizumab treatment, using label-free mass spectrometry and a subset of the changed proteins were verified by parallel reaction monitoring in a new cohort of 20 patients, confirming the majority of observed changes. RESULTS A total of 287 differentially abundant proteins were detected including (i) the decrease of proteins with roles in immunity, such as immunoglobulin heavy constant mu, chitinase-3-like protein 1 and chitotriosidase, (ii) an increase of proteins involved in metabolism, such as lactate dehydrogenase A and B and malate-dehydrogenase cytoplasmic, and (iii) an increase of proteins associated with the central nervous system, including lactadherin and amyloid precursor protein. Comparison with the CSF-PR database provided evidence that natalizumab counters protein changes commonly observed in RRMS. Furthermore, vitamin-D binding protein and apolipoprotein 1 and 2 were unchanged during treatment with natalizumab, implying that these may be involved in disease activity unaffected by natalizumab. CONCLUSIONS Our study revealed that some of the previously suggested biomarkers for MS were affected by the natalizumab treatment while others were not. Proteins not previously suggested as biomarkers were also found affected by the treatment. In sum, the results provide new information on how the natalizumab treatment impacts the CSF proteome of MS patients, and points towards processes affected by the treatment. These findings ought to be explored further to disclose potential novel disease mechanisms and predict treatment responses.
Collapse
Affiliation(s)
- Ragnhild Reehorst Lereim
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
- Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Bergen, Norway
| | - Petra Nytrova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Astrid Guldbrandsen
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
- Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Bergen, Norway
| | - Eva Kubala Havrdova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kjell-Morten Myhr
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Harald Barsnes
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
- Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Bergen, Norway
| | - Frode S. Berven
- Proteomics Unit (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
22
|
Chataway J, Williams T, Li V, Marrie RA, Ontaneda D, Fox RJ. Clinical trials for progressive multiple sclerosis: progress, new lessons learned, and remaining challenges. Lancet Neurol 2024; 23:277-301. [PMID: 38365380 DOI: 10.1016/s1474-4422(24)00027-9] [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: 05/02/2023] [Revised: 10/04/2023] [Accepted: 01/12/2024] [Indexed: 02/18/2024]
Abstract
Despite the success of disease-modifying treatments in relapsing multiple sclerosis, for many individuals living with multiple sclerosis, progressive disability continues to accrue. How to interrupt the complex pathological processes underlying progression remains a daunting and ongoing challenge. Since 2014, several immunomodulatory approaches that have modest but clinically meaningful effects have been approved for the management of progressive multiple sclerosis, primarily for people who have active inflammatory disease. The approval of these drugs required large phase 3 trials that were sufficiently powered to detect meaningful effects on disability. New classes of drug, such as Bruton tyrosine-kinase inhibitors, are coming to the end of their trial stages, several candidate neuroprotective compounds have been successful in phase 2 trials, and innovative approaches to remyelination are now also being explored in clinical trials. Work continues to define intermediate outcomes that can provide results in phase 2 trials more quickly than disability measures, and more efficient trial designs, such as multi-arm multi-stage and futility approaches, are increasingly being used. Collaborations between patient organisations, pharmaceutical companies, and academic researchers will be crucial to ensure that future trials maintain this momentum and generate results that are relevant for people living with progressive multiple sclerosis.
Collapse
Affiliation(s)
- Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK; Medical Research Council Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK; National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK.
| | - Thomas Williams
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Vivien Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Ruth Ann Marrie
- Departments of Medicine and Community Health Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
23
|
Franklin RJM, Bodini B, Goldman SA. Remyelination in the Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041371. [PMID: 38316552 PMCID: PMC10910446 DOI: 10.1101/cshperspect.a041371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
24
|
Riboni-Verri G, Chen BS, McMurran CE, Halliwell GJ, Brown JWL, Coles AJ, Cunniffe NG. Visual outcome measures in clinical trials of remyelinating drugs. BMJ Neurol Open 2024; 6:e000560. [PMID: 38389586 PMCID: PMC10882304 DOI: 10.1136/bmjno-2023-000560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/15/2024] [Indexed: 02/24/2024] Open
Abstract
One of the most promising approaches to delay, prevent or reverse disability progression in multiple sclerosis (MS) is to enhance endogenous remyelination and limit axonal degeneration. In clinical trials of remyelinating drugs, there is a need for reliable, sensitive and clinically relevant outcome measures. The visual pathway, which is frequently affected by MS, provides a unique model system to evaluate remyelination of acute and chronic MS lesions in vivo and non-invasively. In this review, we discuss the different measures that have been used and scrutinise visual outcome measure selection in current and future remyelination trials.
Collapse
Affiliation(s)
- Gioia Riboni-Verri
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Benson S Chen
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Gregory J Halliwell
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - J William L Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Clinical Outcomes Research Unit (CORe), University of Melbourne, Melborune, Melborune, Australia
| | - Alasdair J Coles
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| | - Nick G Cunniffe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Clinical Vision Laboratory, University of Cambridge, Cambridge, UK
| |
Collapse
|
25
|
Goldman SA, Franklin RJM, Osorio J. Stem and progenitor cell-based therapy of myelin disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:283-295. [PMID: 39341659 DOI: 10.1016/b978-0-323-90120-8.00015-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Much of clinical neurology is concerned with diseases of-or involving-the brain's subcortical white matter. Common to these disorders is the loss of myelin, reflecting the elimination or dysfunction of oligodendrocytes and fibrous astrocytes. As such, the introduction of glial progenitor cells, which can give rise to new oligodendrocytes and astrocytes alike, may be a feasible strategy for treating a broad variety of conditions in which white matter loss is causally involved. This review first covers the sourcing and production of human glial progenitor cells, and the preclinical evidence for their efficacy in achieving myelin restoration in vivo. It then discusses both pediatric and adult disease targets for which transplanted glial progenitors may prove of therapeutic value, those challenges that remain in the clinical application of a glial cell replacement strategy, and the clinical endpoints by which the efficacy of this approach may be assessed.
Collapse
Affiliation(s)
- Steven A Goldman
- Sana Biotechnology, Cambridge, MA, United States; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, United States; University of Copenhagen Faculty of Medicine, Copenhagen, Denmark.
| | | | - Joana Osorio
- Sana Biotechnology, Cambridge, MA, United States; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
26
|
Bergeron E, Bouffard MA. Evidence-based management of optic neuritis. Curr Opin Ophthalmol 2024; 35:73-82. [PMID: 37846574 DOI: 10.1097/icu.0000000000001007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
PURPOSE OF REVIEW Optic neuritis can result from several distinct causes, including multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein antibody disease (MOGAD), when not idiopathic. This review discusses evidence-based treatment approaches contingent upon each specific cause of optic neuritis. RECENT FINDINGS Current evidence highlights the need for prompt plasmapheresis as adjunct to intravenous methylprednisolone (IVMP) in patients with NMOSD-associated optic neuritis. Recent advances have included a proliferation of novel disease modifying therapies (DMTs) for long-term management of NMOSD and an understanding of how existing therapeutic options can be leveraged to optimally treat MOGAD. SUMMARY In acute idiopathic or MS-associated optic neuritis, IVMP hastens visual recovery, though it does not substantially affect final visual outcomes. IVMP and adjunctive plasmapheresis are beneficial in the treatment of NMOSD-associated optic neuritis, with a shorter time-to-treatment associated with a higher likelihood of recovery. The natural history of untreated MOGAD-associated optic neuritis is unclear but treatment with IVMP is near-universal given phenotypic similarities with NMOSD. Long-term immunosuppressive therapy is warranted in patients with NMOSD as well as in patients with MOGAD with poor visual recovery or recurrent attacks.
Collapse
Affiliation(s)
- Emilie Bergeron
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
- Département d'ophtalmologie et d'oto-rhino-laryngologie - chirurgie cervico-faciale, Faculté de médecine, Centre Universitaire d'Ophtalmologie, Hôpital du Saint-Sacrement, CHU de Québec-Université Laval, Quebec UO-Recherche-Clinique, Hôpital du Saint-Sacrement, Centre de recherche du CHU de Québec City, Québec City, Quebec, Canada
| | - Marc A Bouffard
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Li M, Liu Q. Inflammatory Demyelinating Diseases of the Central Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 41:171-218. [PMID: 39589715 DOI: 10.1007/978-3-031-69188-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Over the past decades, a large number of immunomodulatory or immunosuppressive treatments have been approved to treat central nervous system (CNS) demyelinating disorders such as multiple sclerosis (MS). Owing to the heterogeneity of patients with CNS demyelinating diseases, there is no clinical treatment that can adequately control all disease subtypes. Although significant progress has been made for relapsing-remitting MS, effective management of the progressive phase of MS has not yet been achieved. This is at least in part caused by our incomplete understanding of the mechanisms driving disease progression, despite our increasing knowledge regarding the underlying cellular and molecular mechanisms. Here, we summarized our current knowledge regarding the mechanisms of CNS demyelinating disorders and their animal models to identify open questions and challenges for existing concepts. We also discussed potential strategies for the future design of immune therapies to treat CNS demyelinating disorders.
Collapse
Affiliation(s)
- Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
28
|
Wilf-Yarkoni A, Feldmann K, Rubarth K, Dorsch EM, Rust R, Urman I, Hellmann MA, Friedman Y, Lotan I, Bialer O, Buenrostro GS, Zimmermann HG, Leutloff C, Schmitz-Hübsch T, Paul F, Asseyer S, Stiebel-Kalish H. Effectiveness of oral prednisone tapering following intravenous methylprednisolone for acute optic neuritis in multiple sclerosis. PLoS One 2023; 18:e0288366. [PMID: 38060614 PMCID: PMC10703274 DOI: 10.1371/journal.pone.0288366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/11/2023] [Indexed: 12/18/2023] Open
Abstract
Acute optic neuritis treatment lacks standardized protocols. The value of oral prednisone taper (OPT) following intravenous methylprednisolone (IVMP) on visual outcome parameters in optic neuritis (ON) has never been explored. In the present retrospective study, we investigated whether OPT after IVMP affects the structural and functional visual outcomes of inaugural clinically isolated syndrome (CIS)- or multiple sclerosis (MS)-ON. Adult patients with acute, inaugural, unilateral CIS- or MS-ON, treated with IVMP in Germany and Israel were stratified into patients treated with IVMP alone-versus IVMP and OPT. Inclusion criteria were age ≥18, CIS or MS diagnosis according to McDonald criteria 2017, available visual acuity (VA) at nadir before treatment initiation and at follow-up ≥5 months, as well as a spectral domain optic coherence tomography (OCT) data scan at follow-up. Exclusion criteria included recurrent ON, concomitant ophthalmological comorbidities, optical coherence tomography (OCT) of insufficient quality and ON-related escalation therapy after IVMP. The structural outcome was defined as the average retinal nerve fiber layer (RNFL) difference between the ON-affected and the unaffected eye, while the functional outcome was defined as the final high-contrast best-corrected VA (HC-BCVA) at follow-up compared to nadir. The comparative analysis was performed using linear regression analysis, adjusted for sex, age, and days-to-treatment. Fifty-one patients met the inclusion criteria (25% male). The mean age was 33.9 (±10.23) years. Twenty-six patients (51%) received OPT following IVMP. There was no difference in nadir HC-BCVA between the groups (0.39 No OPT; 0.49 With OPT, P = 0.36). Adjusted linear regression analysis did not indicate an influence of OPT on RNFL thickness or on HC-BCVA (beta coefficient for RNFL difference in percentages: 0.51, 95%-CI: [-4.58, 5.59], beta coefficient for logMAR: 0.11, 95%; CI [-0.12, 0.35] at follow-up. In conclusion, the addition of OPT to IVMP did not affect RNFL thickness or the final VA in a retrospective cohort of 51 patients with inaugural acute CIS- or MS-ON. The results of this exploratory study are currently being re-examined in a large-scale, demographically diverse, prospective study.
Collapse
Affiliation(s)
- Adi Wilf-Yarkoni
- Neuroimmunology Unit, Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kristina Feldmann
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Emergency and Acute Medicine, Campus Mitte and Virchow, Charité –Universitätsmedizin Berlin, Berlin, Germany
| | - Kerstin Rubarth
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Charité Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| | - Eva-Maria Dorsch
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rebekka Rust
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Ilia Urman
- Neuroimmunology Unit, Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mark A. Hellmann
- Neuroimmunology Unit, Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yitzhak Friedman
- Neuroimmunology Unit, Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itay Lotan
- Neuroimmunology Unit, Department of Neurology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omer Bialer
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
- Neuro-Ophthalmology Unit, Department of Ophthalmology, Rabin Medical Center, Petah Tikva, Israel
| | - Gilberto Solorza Buenrostro
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hanna G. Zimmermann
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carla Leutloff
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tanja Schmitz-Hübsch
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Susanna Asseyer
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
- Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Neuroscience Clinical Research Center, Charité –Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hadas Stiebel-Kalish
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
- Neuro-Ophthalmology Unit, Department of Ophthalmology, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
29
|
Murphy OC, Calabresi PA, Saidha S. Trans-synaptic degeneration as a mechanism of neurodegeneration in multiple sclerosis. Neural Regen Res 2023; 18:2682-2684. [PMID: 37449621 PMCID: PMC10358689 DOI: 10.4103/1673-5374.373661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/10/2023] [Accepted: 02/25/2023] [Indexed: 07/18/2023] Open
Affiliation(s)
- Olwen C. Murphy
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter A. Calabresi
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Shiv Saidha
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
30
|
Panthagani J, O'Donovan C, Aiyegbusi OL, Liu X, Bayliss S, Calvert M, Pesudovs K, Denniston AK, Moore DJ, Braithwaite T. Evaluating patient-reported outcome measures (PROMs) for future clinical trials in adult patients with optic neuritis. Eye (Lond) 2023; 37:3097-3107. [PMID: 36932161 PMCID: PMC10022552 DOI: 10.1038/s41433-023-02478-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/09/2023] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
OBJECTIVE To search for and critically appraise the psychometric quality of patient-reported outcome measures (PROMs) developed or validated in optic neuritis, in order to support high-quality research and care. METHODS We systematically searched MEDLINE(Ovid), Embase(Ovid), PsycINFO(Ovid) and CINAHLPlus(EBSCO), and additional grey literature to November 2021, to identify PROM development or validation studies applicable to optic neuritis associated with any systemic or neurologic disease in adults. We included instruments developed using classic test theory or Rasch analysis approaches. We used established quality criteria to assess content development, validity, reliability, and responsiveness, grading multiple domains from A (high quality) to C (low quality). RESULTS From 3142 screened abstracts we identified five PROM instruments potentially applicable to optic neuritis: three differing versions of the National Eye Institute (NEI)-Visual Function Questionnaire (VFQ): the 51-item VFQ; the 25-item VFQ and a 10-item neuro-ophthalmology supplement; and the Impact of Visual Impairment Scale (IVIS), a constituent of the Multiple Sclerosis Quality of Life Inventory (MSQLI) handbook, derived from the Functional Assessment of Multiple Sclerosis (FAMS). Psychometric appraisal revealed the NEI-VFQ-51 and 10-item neuro module had some relevant content development but weak psychometric development, and the FAMS had stronger psychometric development using Rasch Analysis, but was only somewhat relevant to optic neuritis. We identified no content or psychometric development for IVIS. CONCLUSION There is unmet need for a PROM with strong content and psychometric development applicable to optic neuritis for use in virtual care pathways and clinical trials to support drug marketing authorisation.
Collapse
Affiliation(s)
| | - Charles O'Donovan
- School of Immunology and Microbiology, King's College London, London, UK.
| | - Olalekan Lee Aiyegbusi
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, Birmingham Health Partners for Regulatory Science and Innovation, NIHR Birmingham Biomedical Research Centre, NIHR Applied Research Collaboration West Midlands, and NIHR Birmingham-Oxford Blood and Transplant Research Unit (BTRU) in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Xiaoxuan Liu
- Institute of Inflammation and Ageing, University of Birmingham, University Hospitals Birmingham, Health Data Research UK, London, UK
| | - Susan Bayliss
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Melanie Calvert
- Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, Birmingham Health Partners for Regulatory Science and Innovation, NIHR, Birmingham Biomedical Research Centre, NIHR Surgical Reconstruction and Microbiology Centre, NIHR Applied Research Collaboration West Midlands, and NIHR Birmingham-Oxford Blood and Transplant Research Unit (BTRU) in Precision Transplant and Cellular Therapeutics, University of Birmingham, Birmingham, B15 2TT, UK
| | | | - Alastair K Denniston
- Institute of Inflammation and Ageing, and Centre for Patient Reported Outcomes Research (CPROR), Institute of Applied Health Research, Birmingham Health Partners for Regulatory Science and Innovation, NIHR Birmingham-Oxford Blood and Transplant Research Unit (BTRU) in Precision Transplant and Cellular Therapeutics, University of Birmingham, University Hospitals Birmingham, Health Data Research UK, London, UK
| | - David J Moore
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Tasanee Braithwaite
- School of Immunology and Microbiology, King's College London, and The Medical Eye Unit, Guy's and St Thomas' Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
31
|
Tardo L, Salter A, Truong-Le M, Horton L, Blackburn KM, Sguigna PV. A narrative review of neuro-ophthalmologic disease in African Americans and Hispanics with multiple sclerosis. Ther Adv Chronic Dis 2023; 14:20406223231202645. [PMID: 37790945 PMCID: PMC10542320 DOI: 10.1177/20406223231202645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023] Open
Abstract
Multiple sclerosis (MS) is the most common non-traumatic cause of disability in young people, with vision loss in the disease representing the second largest contributor to disability. In particular, African-American patients with MS are noted to have lower vision than their Caucasian counterparts. In this review, we examine the disparities in eye diseases in the MS population with our gaps in knowledge and discuss the underlying nature of pathological disparities.
Collapse
Affiliation(s)
- Lauren Tardo
- Department of Neurology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-8806, USA
| | - Amber Salter
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie Truong-Le
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lindsay Horton
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kyle M. Blackburn
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peter V. Sguigna
- Department of Neurology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
32
|
Vasileiou ES, Fitzgerald KC. Multiple Sclerosis Pathogenesis and Updates in Targeted Therapeutic Approaches. Curr Allergy Asthma Rep 2023; 23:481-496. [PMID: 37402064 DOI: 10.1007/s11882-023-01102-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2023] [Indexed: 07/05/2023]
Abstract
PURPOSE OF REVIEW In this review, we provide a comprehensive update on current scientific advances and emerging therapeutic approaches in the field of multiple sclerosis. RECENT FINDINGS Multiple sclerosis (MS) is a common disorder characterized by inflammation and degeneration within the central nervous system (CNS). MS is the leading cause of non-traumatic disability in the young adult population. Through ongoing research, an improved understanding of the disease underlying mechanisms and contributing factors has been achieved. As a result, therapeutic advancements and interventions have been developed specifically targeting the inflammatory components that influence disease outcome. Recently, a new type of immunomodulatory treatment, known as Bruton tyrosine kinase (BTK) inhibitors, has surfaced as a promising tool to combat disease outcomes. Additionally, there is a renewed interested in Epstein-Barr virus (EBV) as a major potentiator of MS. Current research efforts are focused on addressing the gaps in our understanding of the pathogenesis of MS, particularly with respect to non-inflammatory drivers. Significant and compelling evidence suggests that the pathogenesis of MS is complex and requires a comprehensive, multilevel intervention strategy. This review aims to provide an overview of MS pathophysiology and highlights the most recent advances in disease-modifying therapies and other therapeutic interventions.
Collapse
Affiliation(s)
- Eleni S Vasileiou
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
33
|
Zhan G, Wang D, Cabezas M, Bai L, Kyle K, Ouyang W, Barnett M, Wang C. Learning from pseudo-labels: deep networks improve consistency in longitudinal brain volume estimation. Front Neurosci 2023; 17:1196087. [PMID: 37483345 PMCID: PMC10358358 DOI: 10.3389/fnins.2023.1196087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Brain atrophy is a critical biomarker of disease progression and treatment response in neurodegenerative diseases such as multiple sclerosis (MS). Confounding factors such as inconsistent imaging acquisitions hamper the accurate measurement of brain atrophy in the clinic. This study aims to develop and validate a robust deep learning model to overcome these challenges; and to evaluate its impact on the measurement of disease progression. Methods Voxel-wise pseudo-atrophy labels were generated using SIENA, a widely adopted tool for the measurement of brain atrophy in MS. Deformation maps were produced for 195 pairs of longitudinal 3D T1 scans from patients with MS. A 3D U-Net, namely DeepBVC, was specifically developed overcome common variances in resolution, signal-to-noise ratio and contrast ratio between baseline and follow up scans. The performance of DeepBVC was compared against SIENA using McLaren test-retest dataset and 233 in-house MS subjects with MRI from multiple time points. Clinical evaluation included disability assessment with the Expanded Disability Status Scale (EDSS) and traditional imaging metrics such as lesion burden. Results For 3 subjects in test-retest experiments, the median percent brain volume change (PBVC) for DeepBVC and SIENA was 0.105 vs. 0.198% (subject 1), 0.061 vs. 0.084% (subject 2), 0.104 vs. 0.408% (subject 3). For testing consistency across multiple time points in individual MS subjects, the mean (± standard deviation) PBVC difference of DeepBVC and SIENA were 0.028% (± 0.145%) and 0.031% (±0.154%), respectively. The linear correlation with baseline T2 lesion volume were r = -0.288 (p < 0.05) and r = -0.249 (p < 0.05) for DeepBVC and SIENA, respectively. There was no significant correlation of disability progression with PBVC as estimated by either method (p = 0.86, p = 0.84). Discussion DeepBVC is a deep learning powered brain volume change estimation method for assessing brain atrophy used T1-weighted images. Compared to SIENA, DeepBVC demonstrates superior performance in reproducibility and in the context of common clinical scan variances such as imaging contrast, voxel resolution, random bias field, and signal-to-noise ratio. Enhanced measurement robustness, automation, and processing speed of DeepBVC indicate its potential for utilisation in both research and clinical environments for monitoring disease progression and, potentially, evaluating treatment effectiveness.
Collapse
Affiliation(s)
- Geng Zhan
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Dongang Wang
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Mariano Cabezas
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
| | - Lei Bai
- Shanghai AI Laboratory, Shanghai, China
| | - Kain Kyle
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | | | - Michael Barnett
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| | - Chenyu Wang
- Brain and Mind Center, The University of Sydney, Sydney, NSW, Australia
- Sydney Neuroimaging Analysis Center, Sydney, NSW, Australia
| |
Collapse
|
34
|
Sorensen PS, Magyari M, Sellebjerg F. An update on combination therapies for multiple sclerosis: where are we now? Expert Rev Neurother 2023; 23:1173-1187. [PMID: 38058171 DOI: 10.1080/14737175.2023.2289572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
INTRODUCTION In theory, combination of two agents, which are suboptimal when given individually, may result in a significant increase in therapeutic effect. Combination therapies have proven particularly effective against infections such as HIV, cancer, and also chronic autoimmune diseases such as rheumatoid arthritis. AREAS COVERED The authors review the literature, searching for randomized placebo-controlled or comparative, double-blind or investigator-blinded clinical trials, not including open label clinical trials, of treatment of multiple sclerosis (MS) with combination therapy or add-on therapy, including trials of induction therapy, trials for prevention of disease activity or worsening, amelioration of adverse effects, and treatment of relapses, and trials to increase remyelination. EXPERT OPINION Combination of two platform therapies (Interferon-beta or glatiramer acetate) was without additional effect. Clinical trials with add-on, often applying repurposed drugs (e.g. simvastatin, atorvastatin, minocycline, estriol, cyclophosphamide, azathioprine, albuterol, vitamin D), have been negative, apart from monthly methylprednisolone that, however, had low tolerability. Combination therapy for neuroprotection/remyelination showed some interesting results, though we are still awaiting results of phase III trials. The results of combination of anti-inflammatory therapies have in general been disappointing. In the future, combination of new effective neuroprotective/remyelinating drugs and highly effective anti-inflammatory treatments may benefit people with MS.
Collapse
Affiliation(s)
- Per Soelberg Sorensen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Melinda Magyari
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Danish Multiple Sclerosis Registry, Department of Neurology, Rigshospitalet, Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Packer D, Fresenko EE, Harrington EP. Remyelination in animal models of multiple sclerosis: finding the elusive grail of regeneration. Front Mol Neurosci 2023; 16:1207007. [PMID: 37448959 PMCID: PMC10338073 DOI: 10.3389/fnmol.2023.1207007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Remyelination biology and the therapeutic potential of restoring myelin sheaths to prevent neurodegeneration and disability in multiple sclerosis (MS) has made considerable gains over the past decade with many regeneration strategies undergoing tested in MS clinical trials. Animal models used to investigate oligodendroglial responses and regeneration of myelin vary considerably in the mechanism of demyelination, involvement of inflammatory cells, neurodegeneration and capacity for remyelination. The investigation of remyelination in the context of aging and an inflammatory environment are of considerable interest for the potential translation to progressive multiple sclerosis. Here we review how remyelination is assessed in mouse models of demyelination, differences and advantages of these models, therapeutic strategies that have emerged and current pro-remyelination clinical trials.
Collapse
|
36
|
Nij Bijvank JA, Hof SN, Prouskas SE, Schoonheim MM, Uitdehaag BMJ, van Rijn LJ, Petzold A. A novel eye-movement impairment in multiple sclerosis indicating widespread cortical damage. Brain 2023; 146:2476-2488. [PMID: 36535900 PMCID: PMC10232247 DOI: 10.1093/brain/awac474] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/04/2023] Open
Abstract
In multiple sclerosis, remyelination trials have yet to deliver success like that achieved for relapse rates with disease course modifying treatment trials. The challenge is to have a clinical, functional outcome measure. Currently, there are none that have been validated, other than visual evoked potentials in optic neuritis. Like vision, quick eye movements (saccades) are heavily dependent on myelination. We proposed that it is possible to extrapolate from demyelination of the medial longitudinal fasciculus in the brainstem to quantitative assessment of cortical networks governing saccadic eye movements in multiple sclerosis. We have developed and validated a double-step saccadic test, which consists of a pair of eye movements towards two stimuli presented in quick succession (the demonstrate eye movement networks with saccades protocol). In this single-centre, cross-sectional cohort study we interrogated the structural and functional relationships of double-step saccades in multiple sclerosis. Data were collected for double-step saccades, cognitive function (extended Rao's Brief Repeatable Battery), disability (Expanded Disability Status Scale) and visual functioning in daily life (National Eye Institute Visual Function Questionnaire). MRI was used to quantify grey matter atrophy and multiple sclerosis lesion load. Multivariable linear regression models were used for analysis of the relationships between double-step saccades and clinical and MRI metrics. We included 209 individuals with multiple sclerosis (mean age 54.3 ± 10.5 years, 58% female, 63% relapsing-remitting multiple sclerosis) and 60 healthy control subjects (mean age 52.1 ± 9.2 years, 53% female). The proportion of correct double-step saccades was significantly reduced in multiple sclerosis (mean 0.29 ± 0.22) compared to controls (0.45 ± 0.22, P < 0.001). Consistent with this, there was a significantly larger double-step dysmetric saccadic error in multiple sclerosis (mean vertical error -1.18 ± 1.20°) compared to controls (-0.54 ± 0.86°, P < 0.001). Impaired double-step saccadic metrics were consistently associated with more severe global and local grey matter atrophy (correct responses-cortical grey matter: β = 0.42, P < 0.001), lesion load (vertical error: β = -0.28, P < 0.001), progressive phenotypes, more severe physical and cognitive impairment (correct responses-information processing: β = 0.46, P < 0.001) and visual functioning. In conclusion, double-step saccades represent a robust metric that revealed a novel eye-movement impairment in individuals with multiple sclerosis. Double-step saccades outperformed other saccadic tasks in their statistical relationship with clinical, cognitive and visual functioning, as well as global and local grey matter atrophy. Double-step saccades should be evaluated longitudinally and tested as a potential novel outcome measure for remyelination trials in multiple sclerosis.
Collapse
Affiliation(s)
- Jenny A Nij Bijvank
- Amsterdam UMC, Department of Neurology, Vrije Universiteit Amsterdam, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
- Amsterdam UMC, Department of Ophthalmology, Vrije Universiteit Amsterdam, Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
| | - Sam N Hof
- Amsterdam UMC, Department of Neurology, Vrije Universiteit Amsterdam, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
| | - Stefanos E Prouskas
- Amsterdam UMC, Department of Anatomy and Neurosciences, Vrije Universiteit Amsterdam, MS Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Amsterdam UMC, Department of Anatomy and Neurosciences, Vrije Universiteit Amsterdam, MS Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
| | - Bernard M J Uitdehaag
- Amsterdam UMC, Department of Neurology, Vrije Universiteit Amsterdam, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
| | - Laurentius J van Rijn
- Amsterdam UMC, Department of Ophthalmology, Vrije Universiteit Amsterdam, Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
- Department of Ophthalmology, Onze Lieve Vrouwe Gasthuis, 1091 AC Amsterdam, The Netherlands
| | - Axel Petzold
- Amsterdam UMC, Department of Neurology, Vrije Universiteit Amsterdam, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
- Amsterdam UMC, Department of Ophthalmology, Vrije Universiteit Amsterdam, Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, 1081 HZ Amsterdam, The Netherlands
- Moorfields Eye Hospital, The National Hospital for Neurology and Neurosurgery and the Queen Square Institute of Neurology, UCL, London EC1V 2PD, UK
| |
Collapse
|
37
|
Klotz L, Antel J, Kuhlmann T. Inflammation in multiple sclerosis: consequences for remyelination and disease progression. Nat Rev Neurol 2023; 19:305-320. [PMID: 37059811 DOI: 10.1038/s41582-023-00801-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/16/2023]
Abstract
Despite the large number of immunomodulatory or immunosuppressive treatments available to treat relapsing-remitting multiple sclerosis (MS), treatment of the progressive phase of the disease has not yet been achieved. This lack of successful treatment approaches is caused by our poor understanding of the mechanisms driving disease progression. Emerging concepts suggest that a combination of persisting focal and diffuse inflammation within the CNS and a gradual failure of compensatory mechanisms, including remyelination, result in disease progression. Therefore, promotion of remyelination presents a promising intervention approach. However, despite our increasing knowledge regarding the cellular and molecular mechanisms regulating remyelination in animal models, therapeutic increases in remyelination remain an unmet need in MS, which suggests that mechanisms of remyelination and remyelination failure differ fundamentally between humans and demyelinating animal models. New and emerging technologies now allow us to investigate the cellular and molecular mechanisms underlying remyelination failure in human tissue samples in an unprecedented way. The aim of this Review is to summarize our current knowledge regarding mechanisms of remyelination and remyelination failure in MS and in animal models of the disease, identify open questions, challenge existing concepts, and discuss strategies to overcome the translational roadblock in the field of remyelination-promoting therapies.
Collapse
Affiliation(s)
- Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada
| | - Tanja Kuhlmann
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Québec, Canada.
- Institute of Neuropathology, University Hospital Münster, Münster, Germany.
| |
Collapse
|
38
|
Oertel FC, Krämer J, Motamedi S, Keihani A, Zimmermann HG, Dimitriou NG, Condor-Montes S, Bereuter C, Cordano C, Abdelhak A, Trip A, Aktas O, Meuth SG, Wiendl H, Ruprecht K, Bellmann-Strobl J, Paul F, Petzold A, Brandt AU, Albrecht P, Green AJ. Visually Evoked Potential as Prognostic Biomarker for Neuroaxonal Damage in Multiple Sclerosis From a Multicenter Longitudinal Cohort. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200092. [PMID: 36878713 PMCID: PMC10026703 DOI: 10.1212/nxi.0000000000200092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND AND OBJECTIVES With the increasing use of visually evoked potentials (VEPs) as quantitative outcome parameters for myelin in clinical trials, an in-depth understanding of longitudinal VEP latency changes and their prognostic potential for subsequent neuronal loss will be required. In this longitudinal multicenter study, we evaluated the association and prognostic potential of VEP latency for retinal neurodegeneration, measured by optical coherence tomography (OCT), in relapsing-remitting MS (RRMS). METHODS We included 293 eyes of 147 patients with RRMS (age [years, median ± SD] 36 ± 10, male sex 35%, F/U [years, median {IQR} 2.1 {1.5-3.9}]): 41 eyes had a history of optic neuritis (ON) ≥6 months before baseline (CHRONIC-ON), and 252 eyes had no history of ON (CHRONIC-NON). P100 latency (VEP), macular combined ganglion cell and inner plexiform layer volume (GCIPL), and peripapillary retinal nerve fiber layer thickness (pRNFL) (OCT) were quantified. RESULTS P100 latency change over the first year predicted subsequent GCIPL loss (36 months) across the entire chronic cohort (p = 0.001) and in (and driven by) the CHRONIC-NON subset (p = 0.019) but not in the CHRONIC-ON subset (p = 0.680). P100 latency and pRNFL were correlated at baseline (CHRONIC-NON p = 0.004, CHRONIC-ON p < 0.001), but change in P100 latency and pRNFL were not correlated. P100 latency did not differ longitudinally between protocols or centers. DISCUSSION VEP in non-ON eyes seems to be a promising marker of demyelination in RRMS and of potential prognostic value for subsequent retinal ganglion cell loss. This study also provides evidence that VEP may be a useful and reliable biomarker for multicenter studies.
Collapse
Affiliation(s)
- Frederike Cosima Oertel
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Julia Krämer
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Seyedamirhosein Motamedi
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Azeen Keihani
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Hanna G Zimmermann
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Nikolaos G Dimitriou
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Shivany Condor-Montes
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Charlotte Bereuter
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Christian Cordano
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Ahmed Abdelhak
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Anand Trip
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Orhan Aktas
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Sven G Meuth
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Heinz Wiendl
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Klemens Ruprecht
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Judith Bellmann-Strobl
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Friedemann Paul
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Axel Petzold
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Alexander U Brandt
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Philipp Albrecht
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF)
| | - Ari J Green
- From the Weill Institute for Neurosciences (F.C.C.O., A.K., S.C.-M., C.C., A.A., A.J.G.), Department of Neurology, University of California San Francisco (UCSF); Experimental and Clinical Research Center (F.C.C.O., S.M., H.G.Z., C.B., J.B.-S., F.P., A.U.B.), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Department of Neurology with Institute of Translational Neurology (J.K., H.W.), University Hospital Münster, Germany; University of California Berkeley (A.K.); Department of Neurology (N.G.D., O.A., S.G.M., P.A.), Medical Faculty, Heinrich-Heine University and University Hospital Düsseldorf, Germany; Department of Neurology (P.A.), Maria Hilf Clinic Moenchengladbach, Germany; Queen Square MS Centre (A.T., A.P.), University College London, UK; Department of Neurology (K.R., F.P.),-Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany; Moorfield's Eye Hospital & The National Hospital for Neurology and Neurosurgery (A.P.); Queen Square Institute of Neurology, University College London, UK; Dutch Neuro-ophthalmology Expertise Centre, Amsterdam, NL; Department of Neurology (A.U.B.), University of California Irvine (UCI); and Department of Ophthalmology (A.J.G.), University of California San Francisco (UCSF).
| |
Collapse
|
39
|
Mey GM, DeSilva TM. Utility of the visual system to monitor neurodegeneration in multiple sclerosis. Front Mol Neurosci 2023; 16:1125115. [PMID: 37063369 PMCID: PMC10090562 DOI: 10.3389/fnmol.2023.1125115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Neurodegeneration occurs early in the multiple sclerosis (MS) disease course and is an important driver of permanent disability. Current immunomodulatory therapies do not directly target neuronal health; thus, there is a critical need to develop neuroprotective strategies in MS. Outcome measures in clinical trials primarily evaluate disease activity and clinical disability scores rather than measures of neurodegeneration. The visual system provides a noninvasive correlate of brain atrophy and neuronal function through structural and functional exams. Furthermore, optic nerve axons and their respective neuronal cell bodies in the retina, in addition to their synaptic input to the thalamus, provide a distinct anatomy to investigate neurodegenerative processes. This review discusses the utility of the visual system as an early output measure of neurodegeneration in MS as well as an important platform to evaluate neuroprotective strategies in preclinical models.
Collapse
Affiliation(s)
| | - Tara M. DeSilva
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
40
|
Zhao X, Jacob C. Mechanisms of Demyelination and Remyelination Strategies for Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24076373. [PMID: 37047344 PMCID: PMC10093908 DOI: 10.3390/ijms24076373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/19/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
All currently licensed medications for multiple sclerosis (MS) target the immune system. Albeit promising preclinical results demonstrated disease amelioration and remyelination enhancement via modulating oligodendrocyte lineage cells, most drug candidates showed only modest or no effects in human clinical trials. This might be due to the fact that remyelination is a sophistically orchestrated process that calls for the interplay between oligodendrocyte lineage cells, neurons, central nervous system (CNS) resident innate immune cells, and peripheral immune infiltrates and that this process may somewhat differ in humans and rodent models used in research. To ensure successful remyelination, the recruitment and activation/repression of each cell type should be regulated in a highly organized spatio–temporal manner. As a result, drug candidates targeting one single pathway or a single cell population have difficulty restoring the optimal microenvironment at lesion sites for remyelination. Therefore, when exploring new drug candidates for MS, it is instrumental to consider not only the effects on all CNS cell populations but also the optimal time of administration during disease progression. In this review, we describe the dysregulated mechanisms in each relevant cell type and the disruption of their coordination as causes of remyelination failure, providing an overview of the complex cell interplay in CNS lesion sites.
Collapse
|
41
|
Nogo-A and LINGO-1: Two Important Targets for Remyelination and Regeneration. Int J Mol Sci 2023; 24:ijms24054479. [PMID: 36901909 PMCID: PMC10003089 DOI: 10.3390/ijms24054479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that causes progressive neurological disability in most patients due to neurodegeneration. Activated immune cells infiltrate the CNS, triggering an inflammatory cascade that leads to demyelination and axonal injury. Non-inflammatory mechanisms are also involved in axonal degeneration, although they are not fully elucidated yet. Current therapies focus on immunosuppression; however, no therapies to promote regeneration, myelin repair, or maintenance are currently available. Two different negative regulators of myelination have been proposed as promising targets to induce remyelination and regeneration, namely the Nogo-A and LINGO-1 proteins. Although Nogo-A was first discovered as a potent neurite outgrowth inhibitor in the CNS, it has emerged as a multifunctional protein. It is involved in numerous developmental processes and is necessary for shaping and later maintaining CNS structure and functionality. However, the growth-restricting properties of Nogo-A have negative effects on CNS injury or disease. LINGO-1 is also an inhibitor of neurite outgrowth, axonal regeneration, oligodendrocyte differentiation, and myelin production. Inhibiting the actions of Nogo-A or LINGO-1 promotes remyelination both in vitro and in vivo, while Nogo-A or LINGO-1 antagonists have been suggested as promising therapeutic approaches for demyelinating diseases. In this review, we focus on these two negative regulators of myelination while also providing an overview of the available data on the effects of Nogo-A and LINGO-1 inhibition on oligodendrocyte differentiation and remyelination.
Collapse
|
42
|
Zhou YN, Jiang L, Zhang Y, Zhou CN, Yang H, He Q, Wang YY, Xiao Q, Huang DJ, Luo YM, Tang Y, Chao FL. Anti-LINGO-1 antibody protects neurons and synapses in the medial prefrontal cortex of APP/PS1 transgenic mice. Neurosci Res 2023:S0168-0102(23)00039-1. [PMID: 36804877 DOI: 10.1016/j.neures.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
The medial prefrontal cortex (mPFC), one of the most vulnerable brain regions in Alzheimer's disease (AD), plays a critical role in cognition. Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein-1 (LINGO-1) negatively affects nerve growth in the central nervous system; however, its role in the pathological damage to the mPFC remains to be studied in AD. In this study, an anti-LINGO-1 antibody was administered to 10-month-old APP/PS1 mice, and behavioral tests, stereological methods, immunohistochemistry and immunofluorescence were used to answer this question. Our results revealed that LINGO-1 was highly expressed in the neurons of the mPFC of AD mice, and the anti-LINGO-1 antibody improved prefrontal cortex-related function and reduced the protein level of LINGO-1, atrophy of the volume, Aβ deposition and massive losses of synapses and neurons in the mPFC of AD mice. Antagonizing LINGO-1 could effectively alleviate the pathological damage in the mPFC of AD mice, which might be an important structural basis for improving prefrontal cortex-related function. Abnormal expression of LINGO-1 in the mPFC may be one of the key targets of AD, and the effect initiated by the anti-LINGO-1 antibody may provide an important basis in the search for drugs for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yu-Ning Zhou
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Experimental Teaching Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Chun-Ni Zhou
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi-Ying Wang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Qian Xiao
- Department of Radioactive Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Du-Juan Huang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan-Min Luo
- Department of Physiology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Yong Tang
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| | - Feng-Lei Chao
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
43
|
Abstract
The multiple sclerosis (MS) neurotherapeutic landscape is rapidly evolving. New disease-modifying therapies (DMTs) with improved efficacy and safety, in addition to an expanding pipeline of agents with novel mechanisms, provide more options for patients with MS. While treatment of MS neuroinflammation is well tailored in the existing DMT armamentarium, concerted efforts are currently underway for identifying neuropathological targets and drug discovery for progressive MS. There is also ongoing research to develop agents for remyelination and neuroprotection. Further insights are needed to guide DMT initiation and sequencing as well as to determine the role of autologous stem cell transplantation in relapsing and progressive MS. This review provides a summary of these updates.
Collapse
Affiliation(s)
- Moein Amin
- Cleveland Clinic, Department of Neurology, Cleveland, OH 44195, USA
| | - Carrie M Hersh
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA
| |
Collapse
|
44
|
Leo H, Kipp M. Remyelination in Multiple Sclerosis: Findings in the Cuprizone Model. Int J Mol Sci 2022; 23:ijms232416093. [PMID: 36555733 PMCID: PMC9783537 DOI: 10.3390/ijms232416093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Markus Kipp
- Correspondence: ; Tel.: +49-(0)-381-494-8400
| |
Collapse
|
45
|
Individual differences in visual evoked potential latency are associated with variance in brain tissue volume in people with multiple sclerosis: An analysis of brain function-structure correlates. Mult Scler Relat Disord 2022; 68:104116. [PMID: 36041331 DOI: 10.1016/j.msard.2022.104116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/16/2022] [Accepted: 08/13/2022] [Indexed: 12/15/2022]
Abstract
Visual evoked potentials (VEP) index visual pathway functioning, and are often used for clinical assessment and as outcome measures in people with multiple sclerosis (PwMS). VEPs may also reflect broader neural disturbances that extend beyond the visual system, but this possibility requires further investigation. In the present study, we examined the hypothesis that delayed latency of the P100 component of the VEP would be associated with broader structural changes in the brain in PwMS. We obtained VEP latency for a standard pattern-reversal checkerboard stimulus paradigm, in addition to Magnetic Resonance Imaging (MRI) measures of whole brain volume (WBV), gray matter volume (GMV), white matter volume (WMV), and T2-weighted fluid attenuated inversion recovery (FLAIR) white matter lesion volume (FLV). Correlation analyses indicated that prolonged VEP latency was significantly associated with lower WBV, GMV, and WMV, and greater FLV. VEP latency remained significantly associated with WBV, GMV, and WMV even after controlling for the variance associated with inter-ocular latency, age, time between VEP and MRI assessments, and other MRI variables. VEP latency delays were most pronounced in PwMS that exhibited low volume in both white and gray matter simultaneously. Furthermore, PwMS that had delayed VEP latency based on a clinically relevant cutoff (VEP latency ≥ 113 ms) in both eyes had lower WBV, GMV, and WMV and greater FLV in comparison to PwMS that had normal VEP latency in one or both eyes. The findings suggest that PwMS that have delayed latency in both eyes may be particularly at risk for exhibiting greater brain atrophy and lesion volume. These analyses also indicate that VEP latency may index combined gray matter and white matter disturbances, and therefore broader network connectivity and efficiency. VEP latency may therefore provide a surrogate marker of broader structural disturbances in the brain in MS.
Collapse
|
46
|
Hill M, Cunniffe N, Franklin R. Seeing is believing: Identifying remyelination in the central nervous system. Curr Opin Pharmacol 2022; 66:102269. [DOI: 10.1016/j.coph.2022.102269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
|
47
|
Nguyen MNL, Zhu C, Kolbe SC, Butzkueven H, White OB, Fielding J, Kilpatrick TJ, Egan GF, Klistorner A, van der Walt A. Early predictors of visual and axonal outcomes after acute optic neuritis. Front Neurol 2022; 13:945034. [PMID: 36158958 PMCID: PMC9493016 DOI: 10.3389/fneur.2022.945034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Background Predicting long-term visual outcomes and axonal loss following acute optic neuritis (ON) is critical for choosing treatment. Predictive models including all clinical and paraclinical measures of optic nerve dysfunction following ON are lacking. Objectives Using a prospective study method, to identify 1 and 3 months predictors of 6 and 12 months visual outcome (low contrast letter acuity 2.5%) and axonal loss [retinal nerve fiber layer thickness and multifocal evoked potential (mfVEP) amplitude] following acute ON. Methods In total, 37 patients of acute ON onset were evaluated within 14 days using between-eye asymmetry of visual acuity, color vision (Ishihara plates), optical coherence tomography, mfVEP, and optic nerve magnetic resonance imaging [magnetic transfer ratio (MTR) and diffusion tensor imaging (DTI)]. Results Visual outcome at 6 and 12 months was best predicted by Ishihara asymmetry at 1 and 3 months following ON onset. Axonal loss at 6 and 12 months was reliably predicted by Ishihara asymmetry at 1 month. Optic nerve MTR and DTI at 3 months post-acute ON could predict axonal loss at 6 and 12 months. Conclusions Simple Ishihara asymmetry testing 1 month after acute ON onset can best predict visual outcome and axonal loss at 6 and 12 months in a clinical or research setting.
Collapse
Affiliation(s)
- Minh N. L. Nguyen
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Chao Zhu
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
| | - Scott C. Kolbe
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Owen B. White
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Joanne Fielding
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
| | | | - Gary F. Egan
- Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| | | | - Anneke van der Walt
- Department of Neurosciences, Monash University, Melbourne, VIC, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| |
Collapse
|
48
|
Ciapă MA, Șalaru DL, Stătescu C, Sascău RA, Bogdănici CM. Optic Neuritis in Multiple Sclerosis—A Review of Molecular Mechanisms Involved in the Degenerative Process. Curr Issues Mol Biol 2022; 44:3959-3979. [PMID: 36135184 PMCID: PMC9497878 DOI: 10.3390/cimb44090272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Multiple sclerosis is a central nervous system inflammatory demyelinating disease with a wide range of clinical symptoms, ocular involvement being frequently marked by the presence of optic neuritis (ON). The emergence and progression of ON in multiple sclerosis is based on various pathophysiological mechanisms, disease progression being secondary to inflammation, demyelination, or axonal degeneration. Early identification of changes associated with axonal degeneration or further investigation of the molecular processes underlying remyelination are current concerns of researchers in the field in view of the associated therapeutic potential. This article aims to review and summarize the scientific literature related to the main molecular mechanisms involved in defining ON as well as to analyze existing data in the literature on remyelination strategies in ON and their impact on long-term prognosis.
Collapse
Affiliation(s)
| | - Delia Lidia Șalaru
- Cardiology Clinic, Institute of Cardiovascular Diseases, 700503 Iași, Romania
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
- Correspondence:
| | - Cristian Stătescu
- Cardiology Clinic, Institute of Cardiovascular Diseases, 700503 Iași, Romania
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Radu Andy Sascău
- Cardiology Clinic, Institute of Cardiovascular Diseases, 700503 Iași, Romania
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Camelia Margareta Bogdănici
- Department of Surgical Specialties (II), University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
- Ophthalmology Clinic, Saint Spiridon Hospital, Iași 700111, Romania
| |
Collapse
|
49
|
Saitakis G, Chwalisz BK. Treatment and Relapse Prevention of Typical and Atypical Optic Neuritis. Int J Mol Sci 2022; 23:9769. [PMID: 36077167 PMCID: PMC9456305 DOI: 10.3390/ijms23179769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Optic neuritis (ON) is an inflammatory condition involving the optic nerve. Several important typical and atypical ON variants are now recognized. Typical ON has a more favorable prognosis; it can be idiopathic or represent an early manifestation of demyelinating diseases, mostly multiple sclerosis (MS). The atypical spectrum includes entities such as antibody-driven ON associated with neuromyelitis optica spectrum disorder (NMOSD) and myelin oligodendrocyte glycoprotein antibody disease (MOGAD), chronic/relapsing inflammatory optic neuropathy (CRION), and sarcoidosis-associated ON. Appropriate and timely diagnosis is essential to rapidly decide on the appropriate treatment, maximize visual recovery, and minimize recurrences. This review paper aims at presenting the currently available state-of-the-art treatment strategies for typical and atypical ON, both in the acute phase and in the long-term. Moreover, emerging therapeutic approaches and novel steps in the direction of achieving remyelination are discussed.
Collapse
Affiliation(s)
- George Saitakis
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Harvard Medical School, Boston, MA 02115, USA
- Athens Eye Hospital, 166 75 Athens, Greece
| | - Bart K. Chwalisz
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, Suite 835, Boston, MA 02114, USA
| |
Collapse
|
50
|
Bierhansl L, Hartung HP, Aktas O, Ruck T, Roden M, Meuth SG. Thinking outside the box: non-canonical targets in multiple sclerosis. Nat Rev Drug Discov 2022; 21:578-600. [PMID: 35668103 PMCID: PMC9169033 DOI: 10.1038/s41573-022-00477-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system that causes demyelination, axonal degeneration and astrogliosis, resulting in progressive neurological disability. Fuelled by an evolving understanding of MS immunopathogenesis, the range of available immunotherapies for clinical use has expanded over the past two decades. However, MS remains an incurable disease and even targeted immunotherapies often fail to control insidious disease progression, indicating the need for new and exceptional therapeutic options beyond the established immunological landscape. In this Review, we highlight such non-canonical targets in preclinical MS research with a focus on five highly promising areas: oligodendrocytes; the blood-brain barrier; metabolites and cellular metabolism; the coagulation system; and tolerance induction. Recent findings in these areas may guide the field towards novel targets for future therapeutic approaches in MS.
Collapse
Affiliation(s)
- Laura Bierhansl
- Department of Neurology, Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- German Center of Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|