Review

Remyelination as a therapeutic target in the treatment of multiple sclerosis

  • Received: 27 March 2025 Revised: 09 October 2025 Accepted: 10 October 2025 Published: 06 November 2025
  • Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) affecting young adults, particularly in North America and Europe, with nearly 2.5 million individuals impacted globally. Characterized by demyelination and neuronal damage, MS involves complex immune-mediated mechanisms. In this review, we focused on the pathophysiological processes of MS, highlighting the roles of T cells, B cells, and proinflammatory cytokines in driving demyelination, which are often the main focus of treatments in the form of immunotherapy. We emphasized remyelination as a key therapeutic target that is necessary for protecting axons and restoring neural function to solve the root problem. Emerging therapies, such as high-dose supplementation with vitamin D and glutathione, appear effective in regulating immune activity and lowering oxidative burden, thus supporting remyelination and neuroprotection. Preclinical models using toxin-induced demyelination have provided valuable insights into the mechanisms of remyelination and identified potential therapeutic targets like LINGO-1 antagonists. Clinical trials, particularly those involving the anti-LINGO-1 monoclonal antibody BIIB033, have demonstrated encouraging results in enhancing remyelination and improving clinical outcomes. LINGO-1 is an inhibitory protein that impairs OPC differentiation. Integrating these innovative approaches into clinical practice could revolutionize MS management by shifting the focus from managing symptoms to promoting CNS repair and long-term recovery. Continued research into the molecular mechanisms of remyelination and the development of targeted therapies is essential for advancing MS treatment and improving the quality of life for patients.

    Citation: Gurprit Kaur Garcha, Mohamed Ahmed. Remyelination as a therapeutic target in the treatment of multiple sclerosis[J]. AIMS Neuroscience, 2025, 12(4): 539-569. doi: 10.3934/Neuroscience.2025027

    Related Papers:

  • Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) affecting young adults, particularly in North America and Europe, with nearly 2.5 million individuals impacted globally. Characterized by demyelination and neuronal damage, MS involves complex immune-mediated mechanisms. In this review, we focused on the pathophysiological processes of MS, highlighting the roles of T cells, B cells, and proinflammatory cytokines in driving demyelination, which are often the main focus of treatments in the form of immunotherapy. We emphasized remyelination as a key therapeutic target that is necessary for protecting axons and restoring neural function to solve the root problem. Emerging therapies, such as high-dose supplementation with vitamin D and glutathione, appear effective in regulating immune activity and lowering oxidative burden, thus supporting remyelination and neuroprotection. Preclinical models using toxin-induced demyelination have provided valuable insights into the mechanisms of remyelination and identified potential therapeutic targets like LINGO-1 antagonists. Clinical trials, particularly those involving the anti-LINGO-1 monoclonal antibody BIIB033, have demonstrated encouraging results in enhancing remyelination and improving clinical outcomes. LINGO-1 is an inhibitory protein that impairs OPC differentiation. Integrating these innovative approaches into clinical practice could revolutionize MS management by shifting the focus from managing symptoms to promoting CNS repair and long-term recovery. Continued research into the molecular mechanisms of remyelination and the development of targeted therapies is essential for advancing MS treatment and improving the quality of life for patients.



    加载中

    Acknowledgments



    The primary author, Gurprit Kaur Garcha, would like to express sincere appreciation to Dr. Mohamed Ahmed for his academic guidance and supervision during the preparation of this manuscript. His role in overseeing the project and providing general advice is gratefully acknowledged.

    Conflict of interest



    The authors declare no conflict of interest.

    Authors' contributions



    Gurprit Kaur Garcha conducted the literature review, analyzed and synthesized the findings, and wrote the entire manuscript. Dr. Mohamed Ahmed provided supervisory support and general academic oversight. All authors have read and approved the final manuscript.

    [1] Miller E, Wachowicz B, Majsterek I (2013) Advances in antioxidative therapy of multiple sclerosis. Curr Med Chem 20: 4720-4730. https://doi.org/10.2174/09298673113209990156
    [2] Lassmann H (2018) Multiple Sclerosis Pathology. Cold Spring Harb Perspect Med 8: a028936. https://doi.org/10.1101/cshperspect.a028936
    [3] Kidd D, Barkhof F, McConnell R, et al. (1999) Cortical lesions in multiple sclerosis. Brain 122: 17-26. https://doi.org/10.1093/brain/122.1.17
    [4] Sommer I, Schachner M (1981) Monoclonal antibodies (O1 to O4) to oligodendrocyte cell surfaces: an immunocytological study in the central nervous system. Dev Biol 83: 311-327. https://doi.org/10.1016/0012-1606(81)90477-2
    [5] Eng LF, Chao FC, Gerstl B, et al. (1968) The maturation of human white matter myelin. Fractionation of the myelin membrane proteins. Biochemistry 7: 4455-4465. https://doi.org/10.1021/bi00852a042
    [6] Smith KJ, Kapoor R, Felts PA (1999) Demyelination: the role of reactive oxygen and nitrogen species. Brain Pathol 9: 69-92. https://doi.org/10.1111/j.1750-3639.1999.tb00212.x
    [7] Foote AK, Blakemore WF (2005) Inflammation stimulates remyelination in areas of chronic demyelination. Brain 128: 528-539. https://doi.org/10.1093/brain/awh417
    [8] Stadelmann C, Wegner C, Brück W (2011) Inflammation, demyelination, and degeneration—recent insights from MS pathology. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1812: 275-282. https://doi.org/10.1016/j.bbadis.2010.07.007
    [9] Fletcher JM, Lalor SJ, Sweeney CM, et al. (2010) T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol 162: 1-11. https://doi.org/10.1111/j.1365-2249.2010.04143.x
    [10] Greter M, Heppner FL, Lemos MP, et al. (2005) Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat Med 11: 328-334. https://doi.org/10.1038/nm1197
    [11] Teh PP, Vasanthakumar A, Kallies A (2015) Development and Function of Effector Regulatory T Cells. Prog Mol Biol Transl Sci 136: 155-174. https://doi.org/10.1016/bs.pmbts.2015.08.005
    [12] Akirav EM, Xu Y, Ruddle NH (2011) Resident B cells regulate thymic expression of myelin oligodendrocyte glycoprotein. J Neuroimmunol 235: 33-39. https://doi.org/10.1016/j.jneuroim.2011.03.013
    [13] Disanto G, Morahan JM, Barnett MH, et al. (2012) The evidence for the role of B cells in multiple sclerosis. Neurology 78: 823-832. https://doi.org/10.1212/WNL.0b013e318249f6f0
    [14] Lee S, Xu L, Shin Y, et al. (2011) A potential link between autoimmunity and neurodegeneration in immune-mediated neurological disease. J Neuroimmunology 235: 56-69. https://doi.org/10.1016/j.jneuroim.2011.02.007
    [15] Larochelle C, Alvarez JI, Prat A (2011) How do immune cells overcome the blood–brain barrier in multiple sclerosis?. FEBS Lett 585: 3770-3780. https://doi.org/10.1016/j.febslet.2011.04.066
    [16] Abulrob A, Sprong H, Van Bergen en Henegouwen P, et al. (2005) The blood-brain barrier transmigrating single domain antibody: mechanisms of transport and antigenic epitopes in human brain endothelial cells. J Neurochem 95: 1201-1214. https://doi.org/10.1111/j.1471-4159.2005.03463.x
    [17] Stallcup WB (1981) The NG2 antigen, a putative lineage marker: immunofluorescent localization in primary cultures of rat brain. Dev Biol 83: 154-165. https://doi.org/10.1016/S0012-1606(81)80018-8
    [18] Eisenbarth GS, Walsh FS, Nirenberg M (1979) Monoclonal antibody to a plasma membrane antigen of neurons. Proc Natl Acad Sci U S A 76: 4913-4917. https://doi.org/10.1073/pnas.76.10.4913
    [19] Bakshi R, Thompson AJ, Rocca MA, et al. (2008) MRI in multiple sclerosis: current status and future prospects. Lancet Neurol 7: 615-625. https://doi.org/10.1016/S1474-4422(08)70137-6
    [20] Dasgupta S, Jana M, Liu X, et al. (2002) Myelin basic protein-primed T cells induce nitric oxide synthase in microglial cells. Implications for multiple sclerosis. J Biol Chem 277: 39327-39333. https://doi.org/10.1074/jbc.M111841200
    [21] Smith KJ, Lassmann H (2002) The role of nitric oxide in multiple sclerosis. Lancet Neurol 1: 232-241. https://doi.org/10.1016/S1474-4422(02)00102-3
    [22] Liñares D, Taconis M, Maña P, et al. (2006) Neuronal nitric oxide synthase plays a key role in CNS demyelination. J Neurosci 26: 12672-12681. https://doi.org/10.1523/JNEUROSCI.0294-06.2006
    [23] Voet S, Prinz M, van Loo G (2019) Microglia in Central Nervous System Inflammation and Multiple Sclerosis Pathology. Trends Mol Med 25: 112-123. https://doi.org/10.1016/j.molmed.2018.11.005
    [24] Boven LA, Van Meurs M, Van Zwam M, et al. (2006) Myelin-laden macrophages are anti-inflammatory, consistent with foam cells in multiple sclerosis. Brain 129: 517-526. https://doi.org/10.1093/brain/awh707
    [25] Traugott U, Reinherz EL, Raine CS (1983) Multiple sclerosis. Distribution of T cells, T-cell subsets, and Ia-positive macrophages in lesions of different ages. J Neuroimmunology 4: 201-221. https://doi.org/10.1016/0165-5728(83)90036-X
    [26] Bitsch A, Bruck W (2002) Differentiation of multiple sclerosis subtypes: implications for treatment. CNS Drugs 16: 405-418. https://doi.org/10.2165/00023210-200216060-00004
    [27] Mehta B, Ramanathan M, Weinstock-Guttman B (2011) Vitamin D and multiple sclerosis: can vitamin D prevent disease progression?. Expert Rev Neurother 11: 469-471. https://doi.org/10.1586/ern.11.44
    [28] Averill-Bates DA (2023) The antioxidant glutathione. Vitam Horm 121: 109-141. https://doi.org/10.1016/bs.vh.2022.09.002
    [29] Podbielska M, Banik NL, Kurowska E, et al. (2013) Myelin Recovery in Multiple Sclerosis: The Challenge of Remyelination. Brain Sci 3: 1282-1324. https://doi.org/10.3390/brainsci3031282
    [30] Raff MC, Miller RH, Noble M (1983) A glial progenitor cell that develops in vitro into an astrocyte or an oligodendrocyte depending on culture medium. Nature 303: 390-396. https://doi.org/10.1038/303390a0
    [31] Pérez-Cerdá F, Sánchez-Gómez MV, Matute C (2015) Pío del Río Hortega and the discovery of the oligodendrocytes. Front Neuroanat 9: 92. https://doi.org/10.3389/fnana.2015.00092
    [32] Zhao X, Jacob C (2023) Mechanisms of Demyelination and Remyelination Strategies for Multiple Sclerosis. Int J Mol Sci 24: 6373. https://doi.org/10.3390/ijms24076373
    [33] Huang JK, Fancy SP, Zhao C, et al. (2011) Myelin regeneration in multiple sclerosis: targeting endogenous stem cells. Neurotherapeutics 8: 650-658. https://doi.org/10.1007/s13311-011-0065-x
    [34] de Castro F, Bribian A (2005) The molecular orchestra of the migration of oligodendrocyte precursors during development. Brain Res Brain Res Rev 49: 227-241. https://doi.org/10.1016/j.brainresrev.2004.12.034
    [35] Faissner S, Gold R (2019) Progressive multiple sclerosis: latest therapeutic developments and future directions. Ther Adv Neurol Disord 12: 1756286419878323. https://doi.org/10.1177/1756286419878323
    [36] Compston A, Coles A (2002) Multiple sclerosis. Lancet 359: 1221-1231. https://doi.org/10.1016/S0140-6736(02)08220-X
    [37] Zhou QH, Fu A, Boado RJ, et al. (2011) Receptor-mediated Aβ amyloid antibody targeting Alzheimer's disease mouse brain. Mol Pharm 8: 280-285. https://doi.org/10.1021/mp1003515
    [38] Damasceno A, Damasceno BP, Cendes F (2014) The clinical impact of cerebellar grey matter pathology in multiple sclerosis. PLoS One 9: e96193. https://doi.org/10.1371/journal.pone.0096193
    [39] Maki T, Liang AC, Miyamoto N, et al. (2013) Mechanisms of oligodendrocyte regeneration from ventricular–subventricular zone derived progenitor cells in white matter diseases. Front Cell Neurosci 7: 275. https://doi.org/10.3389/fncel.2013.00275
    [40] Armstrong RC, Dorn HH, Kufta CV, et al. (1992) Pre-oligodendrocytes from adult human CNS. J Neurosci 12: 1538–1547. https://doi.org/10.1523/JNEUROSCI.12-04-01538.1992
    [41] Sharma R, Sekhon S, Lui F, et al. (2024) White Matter Lesions. StatPearls [Internet] . Treasure Island (FL): StatPearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK562167/
    [42] Simons M, Trajkovic K (2006) Neuron-glia communication in the control of oligodendrocyte function and myelination. Glia 54: 1234-1241.
    [43] McIntyre LJ, Quarles RH, Brady RO (1979) Lectin-binding proteins in central nervous system myelin. Detection of glycoproteins of purified myelin on polyacrylamide gels by [3H] concanavalin A binding. Biochem J 183: 205-212. https://doi.org/10.1042/bj1830205
    [44] Wolswijk G, Balesar R (2003) Changes in the expression and localization of the paranodal protein Caspr on axons in chronic multiple sclerosis. Brain 126: 1638-1649. https://doi.org/10.1093/brain/awg151
    [45] Asakura K, Rodriguez M (1996) Peripheral oral tolerance induces in vivo expansion of Th2 cells and prevents induction of experimental autoimmune encephalomyelitis. J Neuroimmunol 68: 13-23.
    [46] Friedman B, Hockfield S, Black JA, et al. (1989) In situ demonstration of mature oligodendrocytes and their processes: an immunocytochemical study with a new monoclonal antibody, rip. Glia 2: 380-390. https://doi.org/10.1002/glia.440020510
    [47] Waldburger JM, Firestein GS (2010) Regulation of peripheral inflammation by the central nervous system. Curr Rheumatol Rep 12: 370-378. https://doi.org/10.1007/s11926-010-0124-z
    [48] Engelhardt B, Ransohoff RM (2012) Capture, crawl, cross: the T cell code to breach the blood-brain barriers. Trends Immunol 33: 579-589. https://doi.org/10.1016/j.it.2012.07.004
    [49] Alt C, Laschinger M, Engelhardt B (2002) Functional expression of the lymphoid chemokines CCL19 (ELC) and CCL 21 (SLC) at the blood-brain barrier suggests their involvement in G-protein-dependent lymphocyte recruitment into the central nervous system during experimental autoimmune encephalomyelitis. Eur J Immunol 32: 2133-2144. https://doi.org/10.1002/1521-4141(200208)32:8<2133::AID-IMMU2133>3.0.CO;2-W
    [50] Poltorak M, Sadoul R, Keilhauer G, et al. (1987) Myelin-associated glycoprotein, a member of the L2/HNK-1 family of neural cell adhesion molecules, is involved in neuron–oligodendrocyte and oligodendrocyte–oligodendrocyte interaction. J Cell Biol 105: 1893-1899. https://doi.org/10.1083/jcb.105.4.1893
    [51] Harlow DE, Honce JM, Lassmann H (2015) Remyelination therapy in multiple sclerosis. Front Neurol 6: 257. https://doi.org/10.3389/fneur.2015.00257
    [52] Oh LY, Larsen PH, Krekoski CA, et al. (1999) Matrix metalloproteinase 9/gelatinase B is required for process outgrowth by oligodendrocytes. J Neurosci 19: 8464-8475. https://doi.org/10.1523/JNEUROSCI.19-19-08464.1999
    [53] Howell OW, Palser A, Polito A, et al. (2006) Disruption of paranodal axoglial junctions and redistribution of Kv1.2 in chronic multiple sclerosis lesions. Brain 129: 1040-1051.
    [54] Sherman DL, Brophy PJ (2005) Mechanisms of axon ensheathment and myelin growth. Nat Rev Neurosci 6: 683-690. https://doi.org/10.1038/nrn1743
    [55] Olsen JA, Akirav EM (2015) Remyelination in multiple sclerosis: cellular mechanisms and novel therapeutic approaches. J Neurosci Res 93: 687-696. https://doi.org/10.1002/jnr.23493
    [56] Smith KJ, Blakemore WF, McDonald WI (1981) The restoration of conduction by central remyelination. Brain 104: 383-404. https://doi.org/10.1093/brain/104.2.383
    [57] Waxman SG (1977) Conduction in myelinated, unmyelinated, and demyelinated fibers. Arch Neurol 34: 585-589. https://doi.org/10.1001/archneur.1977.00500220019003
    [58] Hayakawa K, Pham LD, Som AT, et al. (2011) Vascular endothelial growth factor regulates the migration of oligodendrocyte precursor cells. J Neurosci 31: 10666-10670. https://doi.org/10.1523/JNEUROSCI.1944-11.2011
    [59] Mathey EK, Derfuss T, Storch MK, et al. (2007) Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med 204: 2363-2372. https://doi.org/10.1084/jem.20071053
    [60] Oliver AR, Lyon GM, Ruddle NH (2003) Rat and human myelin oligodendrocyte glycoproteins induce experimental autoimmune encephalomyelitis by different mechanisms in C57BL/6 mice. J Immunol 171: 462-468. https://doi.org/10.4049/jimmunol.171.1.462
    [61] Irvine KA, Blakemore WF (2008) Remyelination protects axons from demyelination-associated axon degeneration. Brain 131: 1464-1477. https://doi.org/10.1093/brain/awn080
    [62] Lloyd AF, Miron VE (2019) The pro-remyelination properties of microglia in the central nervous system. Nat Rev Neurol 15: 447-458. https://doi.org/10.1038/s41582-019-0184-2
    [63] Pepinsky RB, Arndt JW, Quan C, et al. (2014) Structure of the LINGO-1–Anti-LINGO-1 Li81 Antibody Complex Provides Insights into the Biology of LINGO-1 and the Mechanism of Action of the Antibody Therapy. J Pharmacol Exp Ther 350: 110-123. https://doi.org/10.1124/jpet.113.211771
    [64] Kniesel U, Wolburg H (2000) Tight junctions of the blood-brain barrier. Cell Mol Neurobiol 20: 57-76. https://doi.org/10.1023/A:1006995910836
    [65] Warrington AE, Bieber AJ, Ciric B, et al. (2007) A recombinant human IgM promotes myelin repair after a single, very low dose. J Neurosci Res 85: 967-976. https://doi.org/10.1002/jnr.21217
    [66] Carnegie PR (1971) Amino acid sequence of the encephalitogenic basic protein from human myelin. Biochem J 123: 57-67. https://doi.org/10.1042/bj1230057
    [67] Steinman L (1996) Multiple sclerosis: A coordinated immunological attack against myelin in the central nervous system. Cell 85: 299-302. https://doi.org/10.1016/S0092-8674(00)81107-1
    [68] Howe CL, Bieber AJ, Warrington AE, et al. (2004) Antiapoptotic signaling by a remyelination-promoting human antimyelin antibody. Neurobiol Dis 15: 120-131. https://doi.org/10.1016/j.nbd.2003.09.002
    [69] Jackman N, Ishii A, Bansal R (2009) Oligodendrocyte development and myelin biogenesis: parsing out the roles of glycosphingolipids. Physiology 24: 290-297. https://doi.org/10.1152/physiol.00016.2009
    [70] Iglesias-Rozas JR, Garrosa M (1921) The discovery of oligodendroglia cells by Rio Hortega: his original articles. Clin Neuropathol 31: 437-439.
    [71] Wang Y, Piao JH, Larsen EC, et al. (2011) Migration and remyelination by oligodendrocyte progenitor cells transplanted adjacent to focal areas of spinal cord inflammation. J Neurosci Res 89: 1737-1746. https://doi.org/10.1002/jnr.22716
    [72] Salcedo R, Wasserman K, Young HA, et al. (1999) Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells: In vivo neovascularization induced by stromal-derived factor-1alpha. Am J Pathol 154: 1125-1135. https://doi.org/10.1016/s0002-9440(10)65365-5
    [73] Ardizzone A, Bova V, Casili G, et al. (2023) Role of Basic Fibroblast Growth Factor in Cancer: Biological Activity, Targeted Therapies, and Prognostic Value. Cells 12: 1002. https://doi.org/10.3390/cells12071002
    [74] Berg GJ, Schachner M (1982) Electron-microscopic localization of A2B5 cell surface antigen in monolayer cultures of murine cerebellum and retina. Cell Tissue Res 224: 637-645. https://doi.org/10.1007/BF00213758
    [75] Dietrich J, Noble M, Mayer-Proschel M (2002) Characterization of A2B51 glial precursor cells from cryopreserved human fetal brain progenitor cells. Glia 40: 65-77. https://doi.org/10.1002/glia.10116
    [76] Figarella-Branger D, Colin C, Baeza-Kallee N, et al. (2022) A2B5 Expression in Central Nervous System and Gliomas. Int J Mol Sci 23: 4670. https://doi.org/10.3390/ijms23094670
    [77] Chang A, Nishiyama A, Peterson J, et al. (2000) NG2- positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J Neurosci 20: 6404-6412. https://doi.org/10.1523/JNEUROSCI.20-17-06404.2000
    [78] Bergles DE, Richardson WD (2015) Oligodendrocyte Development and Plasticity. Cold Spring Harb Perspect Biol 8: a020453. https://doi.org/10.1101/cshperspect.a020453
    [79] Miki T, Kaneda M, Iida K, et al. (2013) An anti-sulfatide antibody O4 immunoprecipitates sulfatide rafts including Fyn, Lyn and the G protein α subunit in rat primary immature oligodendrocytes. Glycoconj J 30: 819-823. https://doi.org/10.1007/s10719-013-9487-5
    [80] Palavicini JP, Wang C, Chen L, et al. (2016) Novel molecular insights into the critical role of sulfatide in myelin maintenance/function. J Neurochem 139: 40-54. https://doi.org/10.1111/jnc.13738
    [81] Folch J, Lees M (1951) Proteolipids, a new type of tissue lipoproteins; their isolation from brain. J Biol Chem 191: 807-817. https://doi.org/10.1016/S0021-9258(18)55985-8
    [82] Morell P, Quarles RH (1999) Characteristic Composition of Myelin. Basic Neurochemistry: Molecular, Cellular and Medical Aspects . Philadelphia: Lippincott-Raven.
    [83] Schwob VS, Clark HB, Agrawal D, et al. (1985) Electron microscopic immunocytochemical localization of myelin proteolipid protein and myelin basic protein to oligodendrocytes in rat brain during myelination. J Neurochem 45: 559-571. https://doi.org/10.1111/j.1471-4159.1985.tb04024.x
    [84] Quarles RH (2007) Myelin-associated glycoprotein (MAG): past, present and beyond. J Neurochem 100: 1431-1448. https://doi.org/10.1111/j.1471-4159.2006.04319.x
    [85] Quarles RH, Everly JL, Brady RO (1973) Evidence for the close association of a glycoprotein with myelin in rat brain. J Neurochem 21: 1177-1191. https://doi.org/10.1111/j.1471-4159.1973.tb07573.x
    [86] Kerlero de Rosbo N, Milo R, Lees MB, et al. (1993) Reactivity to myelin antigens in multiple sclerosis. Peripheral blood lymphocytes respond predominantly to myelin oligodendrocyte glycoprotein. J Clin Invest 92: 2602-2608. https://doi.org/10.1172/JCI116875
    [87] Slavin AJ, Johns TG, Orian JM, et al. (1997) Regulation of myelin oligodendrocyte glycoprotein in different species throughout development. Dev Neurosci 19: 69-78. https://doi.org/10.1159/000111187
    [88] Barres BA, Jacobson MD, Schmid R, et al. (1993) Does oligodendrocyte survival depend on axons. Curr Biol 3: 489-497. https://doi.org/10.1016/0960-9822(93)90039-q
    [89] Back SA, Tuohy TM, Chen H, et al. (2005) Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nat Med 11: 966-972. https://doi.org/10.1038/nm1279
    [90] Miron VE, Boyd A, Zhao JW, et al. (2013) M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 16: 1211-1218. https://doi.org/10.1038/nn.3469
    [91] Monge M, Kadiiski D, Jacque CM, et al. (1986) Oligodendroglial expression and deposition of four major myelin constituents in the myelin sheath during development. An in vivo study. Dev Neurosci 8: 222-235. https://doi.org/10.1159/000112255
    [92] Mason JL, Jones JJ, Taniike M, et al. (2000) Mature oligodendrocytes rescued by transplantation form normal myelin in a dysmyelinated host. Brain 123: 765-773.
    [93] Schnaar RL, Lopez PH (2009) Myelin-associated glycoprotein and its axonal receptors. J Neurosci Res 87: 3267-2376. https://doi.org/10.1002/jnr.21992
    [94] Colman DR, Kreibich G, Frey AB, et al. (1982) Synthesis and incorporation of myelin polypeptides into CNS myelin. J Cell Biol 95: 598-608. https://doi.org/10.1083/jcb.95.2.598
    [95] Mi S, Miller RH, Tang W, et al. (2009) Promotion of CNS remyelination by induced differentiation of oligodendrocytes precursor cells. Ann Neurol 65: 304-315. https://doi.org/10.1002/ana.21581
    [96] Franklin RJ, Hinks GL (1999) Understanding CNS remyelination: clues from developmental and regeneration biology. J Neurosci Res 58: 207-213. https://doi.org/10.1002/(SICI)1097-4547(19991015)58:2<207::AID-JNR1>3.0.CO;2-1
    [97] Kosturko LD, Maggipinto MJ, D'Sa C, et al. (2005) The microtubule-associated protein tumor overexpressed gene binds to the RNA trafficking protein heterogeneous nuclear ribonucleoprotein A2. Mol Biol Cell 16: 1938-1947. https://doi.org/10.1091/mbc.e04-08-0709
    [98] Lee J, Gravel M, Zhang R, et al. (2005) Process outgrowth in oligodendrocytes is mediated by CNP, a novel microtubule assembly myelin protein. J Cell Biol 170: 661-673. https://doi.org/10.1083/jcb.200411047
    [99] Ferguson B, Matyszak MK, Esiri MM, et al. (1997) Axonal damage in acute multiple sclerosis lesions. Brain 120: 393-399. https://doi.org/10.1093/brain/120.3.393
    [100] Dirks NL, Meibohm B (2010) Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinetics 49: 633-659. https://doi.org/10.2165/11535960-000000000-00000
    [101] Hauser SL, Bar-Or A, Comi G, et al. (2017) Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis. N Engl J Med 376: 221-234. https://doi.org/10.1056/NEJMoa1601277
    [102] Wiendl H, Hohlfeld R (2002) Therapeutic approaches in multiple sclerosis: lessons from failed and interrupted treatment trials. BioDrugs 16: 183-200. https://doi.org/10.2165/00063030-200216030-00003
    [103] Kania K, Ambrosius W, Kozubski W, et al. (2023) The impact of disease modifying therapies on cognitive functions typically impaired in multiple sclerosis patients: a clinician's review. Front Neurol 14: 1222574. https://doi.org/10.3389/fneur.2023.1222574
    [104] Weiner HL (2009) The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease?. Ann Neurol 65: 239-248. https://doi.org/10.1002/ana.21640
    [105] Lassmann H (2018) Pathogenic mechanisms associated with different clinical courses of multiple sclerosis. Front Immunol 9: 3116. https://doi.org/10.3389/fimmu.2018.03116
    [106] Franklin RJ, Ffrench-Constant C (2008) Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci 9: 839-855. https://doi.org/10.1038/nrn2480
    [107] Franklin RJM, Ffrench-Constant C (2017) Regenerating CNS myelin - from mechanisms to experimental medicines. Nat Rev Neurosci 18: 753-769. https://doi.org/10.1038/nrn.2017.136
    [108] Bezukladova S, Genchi A, Panina-Bordignon P, et al. (2022) Promoting exogenous repair in multiple sclerosis: Myelin regeneration. Curr Opin Neurol 35: 313-318. https://doi.org/10.1097/WCO.0000000000001062
    [109] Green AJ, Gelfand JM, Cree BA, et al. (2017) Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): a randomised, controlled, double-blind, crossover trial. Lancet 390: 2481-2489. https://doi.org/10.1016/S0140-6736(17)32346-2
    [110] Mi S, Pepinsky RB, Cadavid D (2013) Blocking LINGO-1 as a therapy to promote CNS repair: from concept to the clinic. CNS Drugs 27: 493-503. https://doi.org/10.1007/s40263-013-0068-8
    [111] Carlström KE, Zhu K, Ewing E, et al. (2020) Gsta4 controls apoptosis of differentiating adult oligodendrocytes during homeostasis and remyelination via the mitochondria-associated Fas-Casp8-Bid-axis. Nat Commun 11: 4071. https://doi.org/10.1038/s41467-020-17871-5
    [112] Smolders J, Damoiseaux J, Menheere P, et al. (2008) Vitamin D as an immune modulator in multiple sclerosis, a review. J Neuroimmunol 194: 7-17. https://doi.org/10.1016/j.jneuroim.2007.11.014
    [113] Chang A, Staugaitis SM, Dutta R, et al. (2012) Cortical remyelination: a new target for repair therapies in multiple sclerosis. Ann Neurol 72: 918-926. https://doi.org/10.1002/ana.23693
    [114] Lubetzki C, Zalc B, Williams A, et al. (2020) Remyelination in multiple sclerosis: from basic science to clinical translation. Lancet Neurol 19: 678-688. https://doi.org/10.1016/S1474-4422(20)30140-X
    [115] Mosayebi G, Ghazavi A, Ghasami K, et al. (2011) Therapeutic effect of vitamin D3 in multiple sclerosis patients. Immunol Invest 40: 627-639. https://doi.org/10.3109/08820139.2011.573041
    [116] Fernandes de Abreu DA, Eyles D, Féron F (2009) Vitamin D, a neuro-immunomodulator: implications for neurodegenerative and autoimmune diseases. Psychoneuroendocrinology 34 Suppl 1: S265-77. https://doi.org/10.1016/j.psyneuen.2009.05.023
    [117] Hanwell HE, Banwell B (2011) Assessment of evidence for a protective role of vitamin D in multiple sclerosis. Biochim Biophys Acta 1812: 202-212. https://doi.org/10.1016/j.bbadis.2010.07.017
    [118] Harrison SR, Li D, Jeffery LE, et al. (2020) Vitamin D, Autoimmune Disease and Rheumatoid Arthritis. Calcif Tissue Int 106: 58-75. https://doi.org/10.1007/s00223-019-00577-2
    [119] Athanassiou L, Mavragani CP, Koutsilieris M (2022) The Immunomodulatory Properties of Vitamin D. Mediterr J Rheumatol 33: 7-13. https://doi.org/10.31138/mjr.33.1.7
    [120] Matías-Guíu J, Oreja-Guevara C, Matias-Guiu JA, et al. (2018) Vitamin D and remyelination in multiple sclerosis. Neurologia (Engl Ed) 33: 177-186. https://doi.org/10.1016/j.nrl.2016.05.001
    [121] Charier D, Beauchet O, Bell M, et al. (2015) Memantine plus vitamin D prevents axonal degeneration caused by lysed blood. ACS Chem Neurosci 6: 393-397. https://doi.org/10.1021/cn500303k
    [122] Spach KM, Hayes CE (2005) Vitamin D3 confers protection from autoimmune encephalomyelitis only in female mice. J Immunol 175: 4119-4126. https://doi.org/10.4049/jimmunol.175.6.4119
    [123] Holick MF (2007) Vitamin D deficiency. N Engl J Med 357: 266-281. https://doi.org/10.1056/NEJMra070553
    [124] Calabrese G, Morgan B, Riemer J (2017) Mitochondrial Glutathione: Regulation and Functions. Antioxid Redox Signal 27: 1162-1177. https://doi.org/10.1089/ars.2017.7121
    [125] Ferreira B, Mendes F, Osório N, et al. (2013) Glutathione in multiple sclerosis. Br J Biomed Sci 70: 75-79. https://doi.org/10.1080/09674845.2013.11669939
    [126] Ohl K, Tenbrock K, Kipp M (2016) Oxidative stress in multiple sclerosis: Central and peripheral mode of action. Exp Neurol 277: 58-67. https://doi.org/10.1016/j.expneurol.2015.11.010
    [127] Sintzel MB, Rametta M, Reder AT (2018) Vitamin D and Multiple Sclerosis: A Comprehensive Review. Neurol Ther 7: 59-85. https://doi.org/10.1007/s40120-017-0086-4
    [128] Carvalho AN, Lim JL, Nijland PG, et al. (2014) Glutathione in multiple sclerosis: more than just an antioxidant?. Mult Scler 20: 1425-1431. https://doi.org/10.1177/1352458514533400
    [129] Ahmed MS, Shoker A (2010) Vitamin D metabolites; protective versus toxic properties: molecular and cellular perspectives. Nephrol Res Rev 2: 19-26. https://doi.org/10.4081/nr.2010.e5
    [130] Sassi F, Tamone C, D'Amelio P (2018) Vitamin D: Nutrient, Hormone, and Immunomodulator. Nutrients 10: 1656. https://doi.org/10.3390/nu10111656
    [131] Muñoz-Jurado A, Escribano BM, Agüera E, et al. (2022) SARS-CoV-2 infection in multiple sclerosis patients: interaction with treatments, adjuvant therapies, and vaccines against COVID-19. J Neurol 269: 4581-4603. https://doi.org/10.1007/s00415-022-11237-1
    [132] Blakemore WF, Franklin RJ (2008) Remyelination in experimental models of toxin-induced demyelination. Curr Top Microbiol Immunol 318: 193-212. https://doi.org/10.1007/978-3-540-73677-6_8
    [133] Torkildsen O, Brunborg LA, Myhr KM, et al. (2008) The cuprizone model for demyelination. Acta Neurol Scand Suppl 188: 72-76. https://doi.org/10.1111/j.1600-0404.2008.01036.x
    [134] Kipp M, Clarner T, Dang J, et al. (2009) The cuprizone animal model: new insights into an old story. Acta Neuropathol 118: 723-736. https://doi.org/10.1007/s00401-009-0591-3
    [135] Birnbaum JH, Wanner D, Gietl AF, et al. (2018) Oxidative stress and altered mitochondrial protein expression in the absence of amyloid-β and tau pathology in iPSC-derived neurons from sporadic Alzheimer's disease patients. Stem Cell Res 27: 121-130. https://doi.org/10.1016/j.scr.2018.01.019
    [136] Morell P, Barrett CV, Mason JL, et al. (1998) Gene expression in brain during cuprizone-induced demyelination and remyelination. Mol Cell Neurosci 12: 220-227. https://doi.org/10.1006/mcne.1998.0715
    [137] Keough MB, Jensen SK, Yong VW (2015) Experimental demyelination and remyelination of murine spinal cord by focal injection of lysolecithin. J Vis Exp 26: 52679. https://doi.org/10.3791/52679
    [138] Sachs HH, Bercury KK, Popescu DC, et al. (2014) A new model of cuprizone-mediated demyelination/remyelination. ASN Neuro 6: 1759091414551955. https://doi.org/10.1177/1759091414551955
    [139] Tansey FA, Zhang H, Cammer W (1997) Rapid upregulation of the Pi isoform of glutathione-S-transferase in mouse brains after withdrawal of the neurotoxicant, cuprizone. Mol Chem Neuropathol 31: 161-170. https://doi.org/10.1007/BF02815240
    [140] Arnett HA, Mason J, Marino M, et al. (2001) TNF alpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat Neurosci 4: 1116-1122. https://doi.org/10.1038/nn738
    [141] Wergeland S, Torkildsen Ø, Myhr KM, et al. (2011) Dietary vitamin D3 supplements reduce demyelination in the cuprizone model. PLoS One 6: e26262. https://doi.org/10.1371/journal.pone.0026262
    [142] Matsushima GK, Morell P (2001) The neurotoxicant, cuprizone, as a model to study demyelination and remyelination in the central nervous system. Brain Pathol 11: 107-116. https://doi.org/10.1111/j.1750-3639.2001.tb00385.x
    [143] Liu L, Belkadi A, Darnall L, et al. (2010) CXCR2-positive neutrophils are essential for cuprizone-induced demyelination: relevance to multiple sclerosis. Nat Neurosci 13: 319-326. https://doi.org/10.1038/nn.2491
    [144] Skripuletz T, Gudi V, Hackstette D, et al. (2011) De- and remyelination in the CNS white and grey matter induced by cuprizone: the old, the new, and the unexpected. Histol Histopathol 26: 1585-1597. https://doi.org/10.14670/HH-26.1585
    [145] Minagar A (2015) Multiple sclerosis: A mechanistic view. Academic Press.
    [146] Zveik O, Rechtman A, Ganz T, et al. (2024) The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 19: 53. https://doi.org/10.1186/s13024-024-00742-8
    [147] Hall SM (1972) The effect of injections of lysophosphatidyl choline into white matter of the adult mouse spinal cord. J Cell Sci 10: 535-546. https://doi.org/10.1242/jcs.10.2.535
    [148] Kotter MR, Setzu A, Sim FJ, et al. (2001) Macrophage depletion impairs oligodendrocyte remyelination following lysolecithin-induced demyelination. Glia 35: 204-212. https://doi.org/10.1002/glia.1085
    [149] Chu T, Zhang YP, Tian Z, et al. (2019) Dynamic response of microglia/macrophage polarization following demyelination in mice. J Neuroinflammation 16: 188. https://doi.org/10.1186/s12974-019-1586-1
    [150] Sim FJ, Zhao C, Penderis J, et al. (2002) The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J Neurosci 22: 2451-2459. https://doi.org/10.1523/JNEUROSCI.22-07-02451.2002
    [151] Chari DM (2007) Remyelination in multiple sclerosis. Int Rev Neurobiol 79: 589-620. https://doi.org/10.1016/S0074-7742(07)79026-8
    [152] Hiremath MM, Saito Y, Knapp GW, et al. (1998) Microglial/macrophage accumulation during cuprizone-induced demyelination in C57BL/6 mice. J Neuroimmunol 92: 38-49. https://doi.org/10.1016/s0165-5728(98)00168-4
    [153] Plemel JR, Keough MB, Duncan GJ, et al. (2014) Remyelination after spinal cord injury: is it a target for repair?. Prog Neurobiol 117: 54-72. https://doi.org/10.1016/j.pneurobio.2014.02.006
    [154] Chang A, Tourtellotte WW, Rudick R, et al. (2002) Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med 346: 165-173. https://doi.org/10.1056/NEJMoa010994
    [155] Pavelko KD, van Engelen BG, Rodriguez M (1998) Acceleration in the rate of CNS remyelination in lysolecithin-induced demyelination. J Neurosci 18: 2498-2505. https://doi.org/10.1523/JNEUROSCI.18-07-02498
    [156] Mason JL, Jones JJ, Taniike M, et al. (2000) Mature oligodendrocyte apoptosis precedes IGF-1 production and oligodendrocyte progenitor accumulation and differentiation during demyelination/remyelination. J Neurosci Res 61: 251-262. https://doi.org/10.1002/1097-4547(20000801)61:3<251::AID-JNR3>3.0.CO;2-W
    [157] Rae-Grant A, Day GS, Marrie RA, et al. (2018) Practice guideline recommendations summary: disease-modifying therapies for adults with multiple sclerosis: report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 90: 777-788. https://doi.org/10.1212/WNL.0000000000005347
    [158] Neumann B, Baror R, Zhao C, et al. (2019) Metformin Restores CNS Remyelination Capacity by Rejuvenating Aged Stem Cells. Cell Stem Cell 25: 473-485.e8. https://doi.org/10.1016/j.stem.2019.08.015
    [159] Hartley MD, Altowaijri G, Bourdette D (2014) Remyelination and Multiple Sclerosis: Therapeutic Approaches and Challenges. Curr Neurol Neurosci Rep 14: 485. https://doi.org/10.1007/s11910-014-0485-1
    [160] Fancy SP, Chan JR, Baranzini SE, et al. (2011) Myelin regeneration: a recapitulation of development?. Annu Rev Neurosci 34: 21-43. https://doi.org/10.1146/annurev-neuro-061010-113629
    [161] Raine CS (1997) The Norton Lecture: a review of the oligodendrocyte in the multiple sclerosis lesion. J Neuroimmunol 77: 135-152. https://doi.org/10.1016/s0165-5728(97)00073-8
    [162] Kuhlmann T, Miron V, Cui Q, et al. (2008) Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain 131: 1749-1758. https://doi.org/10.1093/brain/awn096
    [163] Mukherjee S, Maxfield FR (2000) Role of membrane organization and membrane domains in endocytic lipid trafficking. Traffic 1: 203-211. https://doi.org/10.1034/j.1600-0854.2000.010302.x
    [164] Mukherjee S, Soe TT, Maxfield FR (1999) Endocytic sorting of lipid analogues differing solely in the chemistry of their hydrophobic tails. J Cell Biol 144: 1271-1284. https://doi.org/10.1083/jcb.144.6.1271
    [165] Dyck SM, Karimi-Abdolrezaee S (2015) Chondroitin sulfate proteoglycans: Key modulators in the developing and pathologic central nervous system. Exp Neurol 269: 169-187. https://doi.org/10.1016/j.expneurol.2015.04.006
    [166] di Penta A, Moreno B, Reix S, et al. (2013) Oxidative stress and proinflammatory cytokines contribute to demyelination and axonal damage in a cerebellar culture model of neuroinflammation. PLoS One 8: e54722. https://doi.org/10.1371/journal.pone.0054722
    [167] Pruvost M, Saadaoui M, Oster H (2021) Oligodendroglial epigenetics, from lineage specification to myelin repair and the role of the microenvironment. Glia 69: 1897-1912. https://doi.org/10.1002/glia.24066
    [168] Kotter MR, Li WW, Zhao C, et al. (2006) Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. J Neurosci 26: 328-332. https://doi.org/10.1523/JNEUROSCI.2615-05.2006
    [169] Zhang X, Gao Y, Zhang S, et al. (2025) Mitochondrial dysfunction in the regulation of aging and aging-related diseases. Cell Commun Signal 23: 290. https://doi.org/10.1186/s12964-025-02308-7
    [170] Zhornitsky S, McKay KA, Metz LM, et al. (2016) Cholesterol and markers of cholesterol turnover in multiple sclerosis: relationship with disease outcomes. Mult Scler Relat Disord 5: 53-65. https://doi.org/10.1016/j.msard.2015.10.005
    [171] Mi S, Miller RH, Lee X, et al. (2005) LINGO-1 negatively regulates myelination by oligodendrocytes. Nat Neurosci 8: 745-751. https://doi.org/10.1038/nn1460
    [172] Heß K, Starost L, Kieran NW, et al. (2020) Lesion stage-dependent causes for impaired remyelination in multiple sclerosis. Acta Neuropathol 140: 359-375. https://doi.org/10.1007/s00401-020-02189-9
    [173] Lucchinetti C, Brück W, Parisi J, et al. (2000) Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol 47: 707-717. https://doi.org/10.1002/1531-8249(200006)47:6<707::aid-ana3>3.0.co;2-q
    [174] Felts PA, Baker TA, Smith KJ (1997) Conduction in segmentally demyelinated mammalian central axons. J Neuroscience 17: 7267-7277. https://doi.org/10.1523/JNEUROSCI.17-19-07267.1997
    [175] Keough MB, Yong VW (2013) Remyelination Therapy for Multiple Sclerosis. Neurotherapeutics 10: 44-54. https://doi.org/10.1007/s13311-012-0152-7
    [176] Pepinsky RB, Shao Z, Ji B, et al. (2011) Exposure levels of anti-LINGO-1 Li81 antibody in the central nervous system and dose-efficacy relationships in rat spinal cord remyelination models after systemic administration. J Pharmacol Exp Ther 339: 519-529. https://doi.org/10.1124/jpet.111.183483
    [177] Mi S, Lee X, Shao Z, et al. (2004) LINGO-1 is a component of the Nogo-66 receptor/p75 signaling complex. Nat Neurosci 7: 221-228. https://doi.org/10.1038/nn1188
    [178] Jepson S, Vought B, Gross CH, et al. (2012) LINGO-1, a transmembrane signaling protein, inhibits oligodendrocyte differentiation and myelination through intercellular self-interactions. J Biol Chem 287: 22184-22195. https://doi.org/10.1074/jbc.M112.366179
    [179] Tran JQ, Rana J, Barkhof F, et al. (2014) Randomized phase I trials of the safety/tolerability of anti-LINGO-1 monoclonal antibody BIIB033. Neurol Neuroimmunol Neuroinflamm 1: e18. https://doi.org/10.1212/NXI.0000000000000018
    [180] Cadavid D, Balcer L, Galetta S, et al. (2017) Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomized, placebo-controlled, phase 2 trial. Lancet Neurol 16: 189-199. https://doi.org/10.1016/S1474-4422(16)30377-5
    [181] Boado RJ, Hui EK, Lu JZ, et al. (2010) Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor- specific IgG fusion protein. J Biotechnol 146: 84-91. https://doi.org/10.1016/j.jbiotec.2010.01.011
    [182] Wang Q, Delva L, Weinreb PH, et al. (2018) Monoclonal antibody exposure in rat and cynomolgus monkey cerebrospinal fluid following systemic administration. Fluids Barriers CNS 15: 10. https://doi.org/10.1186/s12987-018-0093-6
    [183] Aktas O, Ziemssen F, Ziemssen T, et al. (2025) RENEWED: A follow-up study of the opicinumab phase 2 RENEW study in participants with acute optic neuritis. Mult Scler Relat Disord 93: 106185. https://doi.org/10.1016/j.msard.2024.106185
    [184] Petrillo J, Balcer L, Galetta S, et al. (2019) Initial impairment and recovery of vision-related functioning in participants with acute optic neuritis from the RENEW trial of Opicinumab. J Neuroophthalmol 39: 153-160. https://doi.org/10.1097/WNO.0000000000000697
    [185] Petereit HF, Rubbert-Roth A (2009) Rituximab levels in cerebrospinal fluid of patients with neurological autoimmune disorders. Mult Scler 15: 189-192. https://doi.org/10.1177/1352458508098268
    [186] Cadavid D, Mellion M, Hupperts R, et al. (2019) Safety and efficacy of opicinumab in patients with relapsing multiple sclerosis (SYNERGY): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 18: 845-856. https://doi.org/10.1016/S1474-4422(19)30137-1
    [187] Elliott C, Arnold DL, Chen H, et al. (2020) Patterning Chronic Active Demyelination in Slowly Expanding/Evolving White Matter MS Lesions. AJNR Am J Neuroradiol 41: 1584-1591. https://doi.org/10.3174/ajnr.A6742
    [188] Chen JT, Collins DL, Atkins HL, et al. (2008) Magnetization transfer ratio evolution with demyelination and remyelination in multiple sclerosis lesions. Ann Neurol 63: 254-262. https://doi.org/10.1002/ana.21302
    [189] Ahmed Z, Fulton D, Douglas MR (2020) Opicinumab: is it a potential treatment for multiple sclerosis?. Ann Transl Med 8: 892. https://doi.org/10.21037/atm.2020.03.131
    [190] Rodriguez M (2007) Immune mechanisms in the pathogenesis of multiple sclerosis. Brain Pathology 17: 209-214. https://doi.org/10.1111/j.1750-3639.2007.00063.x
    [191] Hauser SL, Bhan AK, Gilles F, et al. (1986) Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions. Ann Neurol 19: 578-587. https://doi.org/10.1002/ana.410190610
    [192] Smith KJ, Blakemore WF, McDonald WI (1979) Central remyelination restores secure conduction. Nature 280: 395-396. https://doi.org/10.1038/280395a0
    [193] Polman CH, Uitdehaag BM (2003) New and emerging treatment options for multiple sclerosis. Lancet Neurol 2: 563-566. https://doi.org/10.1016/s1474-4422(03)00505-2
    [194] de Paula Faria D, de Vries EF, Sijbesma JW, et al. (2014) PET imaging of demyelination and remyelination in the cuprizone mouse model for multiple sclerosis: a comparison between [11C]CIC and [11C]MeDAS. Neuroimage 87: 395-402. https://doi.org/10.1016/j.neuroimage.2013.10.057
    [195] Wergeland S, Myhr KM, Loken-Amsrud KI, et al. (2011) Serum levels of vitamin D related to clinical and MRI markers of disease activity in multiple sclerosis. J Neurol 258: 372-378.
    [196] Neely SA, Williamson JM, Klingseisen A, et al. (2022) Remyelination of the aging central nervous system: Lessons from development and demyelinating disease. Brain 145: 2816-2827. https://doi.org/10.1093/brain/awac038
    [197] Kornek B, Storch MK, Weissert R, et al. (2000) Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. Am J Pathol 157: 267-276. https://doi.org/10.1016/S0002-9440(10)64537-3
    [198] Minagar A (2013) Current and future therapies for multiple sclerosis. Scientifica (Cairo) 2013: 249101. https://doi.org/10.1155/2013/249101
  • Reader Comments
  • © 2025 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(656) PDF downloads(24) Cited by(0)

Article outline

Figures and Tables

Figures(4)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog