Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jun 14.
Published in final edited form as: Curr Opin Neurol. 2008 Jun;21(3):278–283. doi: 10.1097/WCO.0b013e3282fd1875

Myelin Repair Strategies: A Cellular View

Vittorio Gallo 1, Regina Armstrong 2,3
PMCID: PMC2695972  NIHMSID: NIHMS106789  PMID: 18451710

Abstract

Purpose of review

The development of successful myelin repair strategies depends on the detailed knowledge of the cellular and molecular processes underlying demyelination and remyelination in the CNS of animal models and in patients with multiple sclerosis (MS). Based on the complexity of the demyelination and remyelination processes, it should be expected that effective therapeutic approaches will require a combination of strategies for immunomodulation, neuroprotection, and myelin replacement. This brief review highlights recent cellular and molecular findings and indicates that future therapeutic strategies to enhance remyelination may also require combinatorial treatment to accomplish.

Recent findings

The relapsing-remitting course of some forms of MS has typically fueled hope for effective repair of MS lesions if demyelinating activity could be attenuated. Recent findings support the potential of endogenous neural stem cells and progenitor cells to generate remyelinating oligodendrocytes. Importantly, interactions with viable axons and supportive astrocytic responses are required for endogenous immature cells to fulfill their potential remyelinating capacity.

Summary

The research described here will help in identifying the major obstacles to effective remyelination and potential therapeutic targets to guide development of comprehensive approaches for testing in animal models and eventual treatment of patients with MS.

Keywords: Neural progenitors, subventricular zone, oligodendrocytes, astrocytes, demyelination, multiple sclerosis

INTRODUCTION

White matter disorders involving myelin affect millions of individuals around the world and include a wide array of pathologies [1]. Among the most significant and extensively studied is the severe demyelination that occurs in multiple sclerosis (MS) patients. Myelin repair is a desired therapeutic approach for a variety of demyelinating or dysmyelinating disorders that can occur either during development of the CNS or in adulthood. A successful remyelination program would not only promote recovery of action potential propagation in affected axons, but also attenuate further axonal damage in white matter tracts of patients with demyelinating diseases. However, both the demyelination and remyelination processes are complex and involve a sequence of steps that correspond to loss and gain of specific physiological functions. Therefore, it is very likely that only combined interventional approaches that target the multiple rate-limiting steps that prevent successful remyelination will improve clinical outcomes.

Myelin repair therapies are not currently in use for clinical MS or other white matter disorders. However, recent research advances have singled out not only the major cellular players involved in the pathology associated with MS, but also some of the signaling pathways that mediate the cellular damage and the endogenous regenerative response of the CNS. Although two major cell types, oligodendrocytes and neurons, are directly engaged in remyelination, it is clear that astrocytes and microglia are also involved in the inflammatory damage to myelin and oligodendrocytes [2,3]. Last, but not least, it is now widely recognized that an endogenous pool of oligodendrocyte progenitor cells (OPCs) capable of remyelinating axons is present in the adult brain [48**]. Although in disseminated demyelinating diseases, such MS, a therapeutic approach involving OPC transplantation might present several difficulties and might not be effective, grafting of exogenous OPCs has been shown to promote remyelination (9). Therefore, these cells and the molecular mechanisms that either prevent or enhance their maturation to myelinating oligodendrocytes under pathological conditions represent major targets of future therapeutic strategies.

In this review, we will focus on remyelination in the adult CNS, particularly on recent experimental work performed in animal models and on MS tissue. We will discuss some of the major cellular issues currently being debated, in the context of developing effective interventional approaches. Because of space constraints, we will be unable to review in detail other important aspects of myelin repair strategies, including: i) exploiting intracellular pathways (such as transcription factors) for oligodendrocyte regeneration and remyelination; ii) integration of immunosuppressive strategies to shift the immune response toward myelin repair; iii) use of imaging and genetic analysis of MS to tailor myelin repair strategies; iv) developing interventions that may be effective in the context of a varied disease course; and v) analysis of MS in pediatric populations and their relevance for myelin repair strategies. These aspects have been at least in part discussed in other review articles in this issue or elsewhere [3,913].

Endogenous neural progenitors of the subventricular zone and in MS lesions: Cellular targets for myelin repair strategies?

It is now well established that the adult CNS contains an endogenous pool of OPCs that is capable of generating remyelinating oligodendrocytes. These OPCs are found within the subventricular zone (SVZ) at a relatively high concentration, distributed at low abundance throughout the CNS, and often persisting in demyelinated lesions [49,1419**]. Multipotential progenitors capable of myelinating congenitally dysmyelinated brains have also been isolated from the human fetal (ventricular zone) and adult (subcortical white matter) brain [20]. Because of their regenerative potential, substantial effort is being focused on identifying the cellular and developmental properties (e.g. transcription factor “fingerprint”) of adult OPCs, and the molecular pathways that regulate their proliferation, migration and differentiation in response to a demyelinating stimulus. The SVZ has become a center of these studies, since: i) as one of the few neurogenic regions of the adult brain, it comprises a sizeable number of proliferating, undifferentiated OPCs [8**,19**,2126**]; ii) these cells are able to migrate to various regions of the CNS and differentiate into mature glia, including oligodendrocytes [8**,25*,27]; and iii) the SVZ is “activated” after demyelination and its neural progenitor pool is expanded [8**,21]. SVZ activation also occurs in MS tissue, and involves an increase in the density and proliferation of progenitor cells expressing PSA-NCAM, Olig2 and Sox10 [19**]. Importantly, several reports have demonstrated that OPCs can migrate out of the SVZ into different brain regions after demyelination, or under different pathological conditions [8**,21,22,25*,28,29*]. Finally, neural progenitors isolated from the SVZ can be successfully transplanted into the forebrain to replace lost oligodendrocytes and to remyelinate axons [9,30,31].

Based on the cellular and molecular characteristics of the lesion environment, cell-based regenerative therapies that involve SVZ OPCs will have to be optimized by manipulating the inherent properties of these neural progenitors, and/or modifying non-permissive aspects of the lesion environment, to improve OPC generation of remyelinating oligodendrocytes. In particular, exhaustion of the OPC pool [32] and attenuation of its proliferation and differentiation potential with age [18*,3335], together with limited migration capacity [36,37], represent significant impediments that could be overcome by engineering the OPC population [38,39]. However, the long-term stability of the cellular and developmental properties of the engineered OPCs represents a limitation to this approach, and in some cases short-term treatments with exogenous molecules prior to grafting might represent a safer alternative [40]. Finally, turning on the myelination program in OPCs in an unfavorable pathological environment might require mechanisms that are significantly different from reproducing the sequence of molecular and cellular events of normal white matter development. Targeting intrinsic regulators of OPC differentiation, such as Olig1 which plays a prominent functional role during remyelination [41], or by identifying new intrinsic factors [42*,43**], may allow OPCs to more effectively overcome signals in the lesion environment that can inhibit OPC differentiation into remyelinating oligodendrocytes.

The dynamic nature of the cellular changes occurring in the SVZ contrasts with the cellular pathology of the periventricular white matter (WM) as a crucial site of MS lesions and area of poor remyelination. In fact, although WM OPCs have been clearly identified in MS lesions, this population displays only limited proliferative and remyelinating capacity as progression of the disease occurs [5,32,4446]. Furthermore, in WM, the density of PSA-NCAM+ progenitors is much higher in active and chronic active MS lesions than in chronic silent lesions [19**]. In active lesions, these undifferentiated progenitors were found most frequently in periventricular WM [19**]. In order to promote oligodendrogenesis and remyelination from progenitor cells present in the lesions, it will be important to define the role of intrinsic and extrinsic mechanisms that underlie the distinction between neural progenitor characteristics in active vs. silent lesions of the WM, as well as in the activated vs. normal SVZ. Furthermore, it will also have to be established whether intracellular transcription factor programs that promote progenitor maturation to myelinating oligodendrocytes are differentially activated in neural progenitors in distinct types of MS lesions.

How to integrate strategies to prevent axon damage or promote axon regeneration with strategies to repair myelin

Remyelination and functional recovery cannot occur without maintaining axonal integrity, including structural (e.g. nodes of Ranvier) and functional (e.g. clustering of Na+ and K+ channels for action potential propagation) properties of the axon that are essential for rapid propagation of action potentials [47]. In MS or other demyelinating disorders, axonal damage can either occur primarily in the acute inflammatory phase of the disease, or as a secondary result of chronic demyelination [4850]. At least two sets of independent observations argue for a direct relationship between the extent of myelination and maintenance of axonal integrity and function. First, mutant mice carrying specific deletions of oligodendrocyte-specific genes display axonal degeneration [51,52], indicating that oligodendrocytes are necessary not only to maintain myelinated tracts, but also axonal integrity. Second, in animal models of demyelination/remyelination, grafting of stem cells [53], oligodendroglial lineage cells [54,55**], Schwann cells [56], or olfactory ensheating cells [57,58] not only promotes remyelination, but also improves electrical conduction along corresponding fiber tracts.

In axons, the molecular structures of the nodes of Ranvier and the paranodes depend on selective interactions between several crucial proteins and the clustering of specific ionic channels [47]. Analysis of MS tissue demonstrated disruption of the clustering of nodal, paranodal and juxtaparanodal proteins, including sodium channels. Conversely, remyelinated lesions displayed normal aggregation of these proteins [59]. These results, together with the findings from animal models discussed above, suggest that promoting the formation of paranodal and juxtaparanodal structures after demyelination might not only enhance axonal recovery, but may also accelerate remyelination before irreversible axonal damage and loss of function have occurred. Among possible molecular targets, two isoforms of neurofascin might play a pivotal role, since these proteins are required for clustering of sodium channels at the node and for the proper assembly of paranodal structures [60]. Furthermore, neurofascin localization is altered in paranodal structures in MS lesions, which appears to occur before demyelination begins [61].

In order to integrate strategies to prevent axon damage - or promote axon regeneration - with strategies to repair myelin, inhibitors of remyelination expressed by axons and OPCs, such as PSA-NCAM [62,63] and LINGO-1 [64**,65*], will also have to be targeted. Regulation of expression of these inhibitors under pathological conditions is still undefined, but it is likely to represent a significant impediment to remyelination and recovery of axonal function. Future therapeutic approaches might be directed at modulating expression of these inhibitors through specific upstream regulatory pathways [65*].

Increased expression of neurotrophic factors in MS lesions may indicate a compensatory effect and a putative therapeutic target to enhance functional recovery. In MS tissue, the ciliary neurotrophic factor (CNTF) pathway is activated in cortical neurons, possibly as a neuroprotective response [66*]. Brain-derived neurotrophic factor (BDNF) can be secreted by both neurons and immune cells in MS brains. A complicated interaction between BDNF levels and MS pathology is suggested by correlations of immune cell BDNF secretion, which can be activity dependent, and imaging parameters of white and gray matter volume [67,68]. Finally, immunomodulatory treatments themselves can regulate neurotrophin expression and/or directly interact with neurotrophic signaling pathways [69], suggesting that immune cell-mediated delivery of neurotrophic factors should be taken into consideration in designing repair strategies in MS.

Targeting astrocytes as pivotal cellular elements of damage and repair processes in demyelinating disease

Astrocytes are a prominent feature of MS lesions. Importantly, different astrocytic characteristics have been implicated in processes that add to the ongoing damage and/or perturb repair, as well as in processes that delimit damaged tissue and promote repair [70,71]. This dichotomy of astrocytic effects has the potential to significantly influence the extent to which the lesion environment is supportive versus prohibitive to OPC recruitment and differentiation, oligodendrocyte survival, and remyelination. The pathological features of an MS lesion may evolve throughout the disease course from an early stage with active demyelination and hypertrophic or “reactive” astrocytes to a chronic inactive phase as a hypocellular plaque with fibrillary gliosis [72]. Inflammation associated with active demyelination corresponds with an environment that is conducive for remyelination [73, 74]. Reactive astrocytes in active lesions may secrete growth factors, cytokines, and chemokines that contribute to the inflammatory response but also stimulate OPC proliferation and recruitment into lesion areas. In contrast, chronic plaques have a central region of demyelinated axons, usually with a reduced axonal density, and an environment with abnormal extracellular matrix composition and a dense network of astrocytic processes forming a glial scar.

Therapeutic approaches that modify the environment of chronic MS lesions may be able to optimize the repair capacity of endogenous cells (see section on SVZ progenitors above) as well as improve the potential of transplanted cells to remyelinate effectively. Astrocytes in non-remyelinating areas associated with the glial scar express: i) vimentin, nestin, PSA-NCAM, FGF2 and NGF [75]; ii) extracellular matrix molecules, such as tenascin family members, as well as Jagged-1 and high molecular weight hyaluronan that inhibit OPC migration and differentiation, respectively [7678]; and a variety of chondroitin sulfate proteoglycans [79]. An attractive means to modify the lesion environment may be to exploit signaling pathways that regulate astrocytic transition from an early reactive state that supports repair processes to a glial scar phenotype that expresses signals that inhibit regeneration. Alternatively, modification of the lesion environment can be targeted to a specific non-permissive molecule of the glial scar. Both approaches have been addressed in similar studies of axonal regeneration, and to some extent remyelination, relative to glial scar formation as a consequence of traumatic spinal cord injury. To move forward in approaches to modify the lesion environment in demyelinating diseases, more complex analyses of the astrocytic response will be needed to consider the astrocytic characteristics relative to remyelination processes. For example, bone morphogenic protein (BMP) family members have increased expression in demyelinated lesions and induce astrogliosis while inhibiting OPC differentiation into oligodendrocytes [8082]. BMP and other potential pathways that may regulate glial scar formation or specific non-permissive signals will be of interest as therapeutic targets if shown to impact the rate or extent of remyelination, especially in the context of chronic lesions.

CONCLUSION

The variability in the clinical symptoms observed in MS patients is matched by significant differences found in MS lesions at the pathological and cellular levels. In many instances, a remyelination program appears to be successfully activated, but in other cases it is severely impaired [83**,84*]. This striking inconsistency is most likely due to the complexity of the cellular interactions that occur during myelination and demyelination, and the fragile balance between cell damage and survival in a pathological milieu. Significant advances have been made in our understanding of some of the crucial cellular and molecular steps required for successful axonal myelination and remyelination. Oligodendrocyte regeneration, preservation of axonal functions and promoting changes in astrocytes to a “permissive” phenotype - together with modulation of the immune response – appear to be the most promising future approaches to enhance remyelination in demyelinating diseases.

graphic file with name nihms106789f1.jpg

The cellular and molecular components of chronically demyelinated lesions present obstacles to remyelination that may also serve as points of intervention to target therapeutically to enhance repair capacity. During early stages of demyelination, inflammatory processes can cause varying degrees of axonal damage. Axonal atrophy and loss can continue during chronic stages, which over time can accumulate and contribute to disability. Oligodendrocyte loss varies among different classes of MS pathology and may be worsened if newly generated oligodendrocytes have poor survival in the environment of chronic lesions. OP cells may be present in germinal zones, such as the subventricular zone (SVZ) adjacent to the lateral ventricles (LV), and distributed within normal tissues but appear to be depleted from areas of chronic demyelination. Astrocytes are activated in response to demyelination and contribute to the non-premissive environment of chonic plaques by scar formation that is expected to impair OP responses and axon outgrowth.

ACKNOWLEDGEMENTS

The authors of this review were supported by National Institutes of Health grants R01NS045702 and R01NS1056427 (VG), R01NS39293 (RCA) and MRDDRC P30HD40677 (VG), by grants RG4019 (VG) and RG3515 (RCA) from the National Multiple Sclerosis Society, and by Department of Defense grant F170TP (RCA). VG was also partially supported by the European Leukodystrophy Association.

Abbreviations

BDNF

brain-derived neurotrophic factor

BMP

bone morphogenic protein

CNS

central nervous system

CNTF

ciliary neurotrophic factor

EGF

epidermal growth factor

EGFR

epidermal growth factor receptor

FGF

fibroblast growth factor

MS

multiple sclerosis

NGF

nerve growth factor

OPC

oligodendrocyte progenitor cell

PSA-NCAM

polysialylated neural cell adhesion molecule

SVZ

subventricular zone

WM

white matter

REFERENCES AND RECOMMENDED READING

  • 1.Gallo V, de Vellis J. White matter disorders. Ment. Ret. Dev. Disabil. Res. Rev. 2006:12. doi: 10.1002/mrdd.20109. [DOI] [PubMed] [Google Scholar]
  • 2.Zhang SC, Goetz BD, Carré JL, Duncan ID. Reactive microglia in dysmyelination and demyelination. Glia. 2001;34:101–109. doi: 10.1002/glia.1045. [DOI] [PubMed] [Google Scholar]
  • 3.Frederick TJ, Miller SD. Future of multiple sclerosis therapy: combining antigen-specific immunotherapy with strategies to promote myelin repair. Future Neurol. 2006;1:489–503. [Google Scholar]
  • 4.Gensert JM, Goldman JE. Endogenous progenitors remyelinate demyelinated axons in the adult CNS. Neuron. 1997;19:197–203. doi: 10.1016/s0896-6273(00)80359-1. [DOI] [PubMed] [Google Scholar]
  • 5.Chang A, Nishiyama A, Peterson J, et al. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J. Neurosci. 2000;20:6404–6412. doi: 10.1523/JNEUROSCI.20-17-06404.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Levine JM, Reynolds R, Fawcett JW. The oligodendrocyte precursor cell in health and disease. Trends Neurosci. 2001;24:39–47. doi: 10.1016/s0166-2236(00)01691-x. [DOI] [PubMed] [Google Scholar]
  • 7.Polito A, Reynolds R. NG2-expressing cells as oligodendrocyte progenitors in the normal and demyelinated adult central nervous system. J. Anat. 2005;207:707–716. doi: 10.1111/j.1469-7580.2005.00454.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Aguirre A, Dupree JL, Mangin JM, Gallo V. A functional role for EGFR signaling in myelination and remyelination. Nat. Neurosci. 2007;10:990–1002. doi: 10.1038/nn1938.. ** EGFR signaling regulates SVZ generation of OP cells and remyelinating oligodendrocytes with a demonstrated functional effect on axon conduction in the corpus callosum.
  • 9.Halfpenny C, Benn T, Scolding N. Cell transplantation, myelin repair and multiple sclerosis. The Lancet, Neurology. 2002;1:31–40. doi: 10.1016/s1474-4422(02)00004-2. [DOI] [PubMed] [Google Scholar]
  • 10.Dubois–Dalcq M, ffrench-Constant C, Franklin RJM. Enhancing central nervous system remyelination in multiple sclerosis. Neuron. 2005;48:9–12. doi: 10.1016/j.neuron.2005.09.004. [DOI] [PubMed] [Google Scholar]
  • 11.Armstrong RC. Growth factor regulation of remyelination: Behind the growing interest in endogenous cell repair of the CNS. Future Neurol. 2007;2:689–697. doi: 10.2217/14796708.2.6.689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Miller RH, Mi S. Dissecting myelination. Nat. Neurosci. 2007;10:1351–1354. doi: 10.1038/nn1995. [DOI] [PubMed] [Google Scholar]
  • 13.Radtke C, Spies M, Sasaki M, Vogt PM, et al. Demyelinating diseases and potential repair strategies. Int. J. Dev. Neurosci. 2007;25:149–153. doi: 10.1016/j.ijdevneu.2007.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Armstrong RC, Dorn HH, Kufta CV, Friedman E, Dubois-Dalcq ME. Pre-oligodendrocytes from adult human CNS. J. Neurosci. 1992;12:1538–1547. doi: 10.1523/JNEUROSCI.12-04-01538.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Armstrong RC, Le TQ, Flint NC, Vana AC, Zhou YX. Endogenous cell repair of chronic demyelination. J. Neuropathol. Exp. Neurol. 2006;65:245–56. doi: 10.1097/01.jnen.0000205142.08716.7e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Dawson MR, Levine JM, Reynolds R. NG2-expressing cells in the central nervous system: are they oligodendroglial progenitors? J. Neurosci. Res. 2000;61:471–479. doi: 10.1002/1097-4547(20000901)61:5<471::AID-JNR1>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 17.Marshall CA, Suzuki SO, Goldman JE. Gliogenic and neurogenic progenitors of the subventricular zone: who are they, where did they come from, and where are they going? Glia. 2003;43:52–61. doi: 10.1002/glia.10213. [DOI] [PubMed] [Google Scholar]
  • 18. Jablonska B, Aguirre A, Vandenbosch R, et al. Cdk2 is critical for proliferation and self-renewal of neural progenitor cells in the adult subventricular zone. J Cell Biol. 2007;6:1231–1245. doi: 10.1083/jcb.200702031.. * A functional demonstration that the function of Cdk2 in SVZ progenitor proliferation is developmentally regulated, as Cdk4 compensates for Cdk2 during the first two postnatal weeks, but not in the adult brain.
  • 19. Nait-Oumesmar B, Picard-Riera N, Kerninon C, et al. Activation of the subventricular zone in multiple sclerosis: evidence for early glial progenitors. Proc. Natl. Acad. Sci. (USA) 2007;104:4694–4699. doi: 10.1073/pnas.0606835104.. ** MS lesions of the periventricular white matter activate the cells of the adjacent SVZ, which may indicate a role for adult human SVZ cells in CNS repair, including remyelination.
  • 20.Windrem MS, Nunes MC, Rashbaum WK, et al. Fetal and adult human oligodendrocyte progenitor cell isolates myelinate the congenitally dysmyelinated brain. Nat. Med. 2004;10:93–97. doi: 10.1038/nm974. [DOI] [PubMed] [Google Scholar]
  • 21.Nait-Oumesmar B, Decker L, Lachapelle F, et al. Progenitor cells of the adult mouse subventricular zone proliferate, migrate and differentiate into oligodendrocytes after demyelination. Eur. J. Neurosci. 1999:4357–4366. doi: 10.1046/j.1460-9568.1999.00873.x. [DOI] [PubMed] [Google Scholar]
  • 22.Picard-Riera N, Decker L, Delarasse C, et al. Experimental autoimmune encephalomyelitis mobilizes neural progenitors from the subventricular zone to undergo oligodendrogenesis in adult mice.Proc. Natl. Acad. Sci. (USA) 2002;99:13211–13216. doi: 10.1073/pnas.192314199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Aguirre AA, Chittajallu R, Belachew S, Gallo V. NG2-expressing cells in the subventricular zone are type C-like cells and contribute to interneuron generation in the postnatal hippocampus. J. Cell Biol. 2004;165:575–589. doi: 10.1083/jcb.200311141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Parras CM, Galli R, Britz O, et al. Mash 1 specifies neurons and oligodendrocytes in the postnatal brain. EMBO J. 2004;23:4495–4505. doi: 10.1038/sj.emboj.7600447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Menn B, Garcia-Verdugo J, Yaschine C, et al. Origin of oligodendrocytes in the subventricular zone of the adult brain. J. Neurosci. 2006;26:7907–7918. doi: 10.1523/JNEUROSCI.1299-06.2006.. * SVZ astrocytes (type B cells) give rise to a small number of OP cells and mature oligodendrocytes in the non-lesioned adult rodent CNS and the number of oligodendrocytes generated increases several fold after a demyelinating lesion of the corpus callosum.
  • 26. Parras CM, Hunt C, Sugimori M, et al. The proneural gene Mash 1 specifies an early population of telencephalic oligodendrocytes. J. Neurosci. 2007;27:4233–4242. doi: 10.1523/JNEUROSCI.0126-07.2007.. ** Direct and functional demonstration that Mash1 is involved in the generation of a subset of oligodendrocyte precursors in the developing ventral forebrain by cooperating with Olig2.
  • 27.Levison SW, Goldman JE. Both oligodendrocytes and astrocytes develop from progenitors in the subventricular zone of postnatal rat forebrain. Neuron. 1993;10:201–212. doi: 10.1016/0896-6273(93)90311-e. [DOI] [PubMed] [Google Scholar]
  • 28.Romanko MJ, Rola R, Fike JR, et al. Roles of the mammalian subventricular zone in cell replacement after brain injury. Prog. Neurobiol. 2004;74:77–99. doi: 10.1016/j.pneurobio.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 29. Gordon RJ, Tattersfield AS, Vazey EM, et al. Temporal profile of subventricular zone progenitor cell migration following quinolinic acid-induced striatal cell loss. Neuroscience. 2007;146:1704–1718. doi: 10.1016/j.neuroscience.2007.03.011.. * Demonstration that directed migration of SVZ-derived progenitor cells and neuroblast differentiation in response to quinolinic acid lesioning of the striatum is acute and transient.
  • 30.Vitry S, Avellana-Adalid V, Lachapelle F, et al. Migration and multipotentiality of PSA-NCM+ neural precursors transplanted in the developing brain. Mol. Cell. Neurosci. 2001;17:983–1000. doi: 10.1006/mcne.2001.0987. [DOI] [PubMed] [Google Scholar]
  • 31.Lachapelle F, Avellana-Adalid, Nait-Oumesmar B, Baron-Van Evercooren A. Fibroblast growth factor-2 (FGF-2) and Platelet-derived growth factor AB (PDGFAB) promote adult SVZ-derived oligodendrogenesis in vivo. Mol. Cell. Neurosci. 2002;17:390–403. doi: 10.1006/mcne.2002.1124. [DOI] [PubMed] [Google Scholar]
  • 32.Mason JL, Toews A, Hostettler JD, et al. Oligodendrocytes and progenitors become progressively depleted with chronically demyelinated lesions. Am. J. Path. 2004;164:1673–1682. doi: 10.1016/S0002-9440(10)63726-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wolswijk G, Noble M. Identification of an adult-specific glial progenitor cell. Development. 1989;105:387–400. doi: 10.1242/dev.105.2.387. [DOI] [PubMed] [Google Scholar]
  • 34.Sim F, Zhao C, Penderis J, Franklin R. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J. Neurosci. 2002;22:2451–2459. doi: 10.1523/JNEUROSCI.22-07-02451.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fancy SPJ, Zhao C, Franklin R. Increased expression of Nkx2.2 and Olig 2 identifies reactive oligodendrocyte progenitor cells responding to demyelination in the adult CNS. Mol. Cell. Neurosci. 2004;27:247–254. doi: 10.1016/j.mcn.2004.06.015. [DOI] [PubMed] [Google Scholar]
  • 36.Franklin RJ, Blakemore WF. To what extent is oligodendrocyte progenitor migration a limiting factor in the remyelination of multiple sclerosis lesions? Mul.Scler. 1997;3:84–87. doi: 10.1177/135245859700300205. [DOI] [PubMed] [Google Scholar]
  • 37.De Castro F, Bribián A. The molecular orchestra of the migration of oligodendrocyte precursors during development. Brain Res. Brain Res. Rev. 2005;49:227–241. doi: 10.1016/j.brainresrev.2004.12.034. [DOI] [PubMed] [Google Scholar]
  • 38.Franceschini I, Vitry S, Padilla F, et al. Migrating and myelinating potential of neural precursors engineered to overexpress PSA-NCAM. Mol. Cell. Neurosci. 2004;27:151–162. doi: 10.1016/j.mcn.2004.05.006. [DOI] [PubMed] [Google Scholar]
  • 39.Glaser T, Perez-Bouza A, Klein K, Brüstle O. Generation of purified oligodendrocyte progenitors from embryonic stem cells. FASEB J. 2005;19:112–114. doi: 10.1096/fj.04-1931fje. [DOI] [PubMed] [Google Scholar]
  • 40.Torregrossa P, Buhl L, Bancila M, et al. Selection of poly-alpha 2,8-sialic acid mimotopes from a random phage peptide library and analysis of their bioactivity. J. Biol. Chem. 2004;279:30707–30714. doi: 10.1074/jbc.M403935200. [DOI] [PubMed] [Google Scholar]
  • 41.Arnett HA, Fancy SPJ, Alberta JA, et al. bHLH transcription factor olig1 is required to repair demyelinated lesions in the CNS. Science. 2004;306:2111–2115. doi: 10.1126/science.1103709. [DOI] [PubMed] [Google Scholar]
  • 42. Sohn J, Natale J, Chew LJ, et al. Identification of Sox17 as a transcription factor that regulates oligodendrocyte development. J. Neurosci. 2006;26:9722–9735. doi: 10.1523/JNEUROSCI.1716-06.2006.. * Demonstration that Sox17 expression is developmentally regulated in the oligodendrocyte lineage and that Sox17 promotes oligodendrocyte progenitor differentiation.
  • 43. He Y, Dupree J, Wang J, et al. The transcription factor Yin Yang 1 is essential for oligodendrocyte progenitor differentiation. Neuron. 2007;55:217–230. doi: 10.1016/j.neuron.2007.06.029.. ** The transcription factor Yin Yang 1 regulates the transition of oligodendrocyte progenitors from cell cycle exit to differentiation, and acts as a lineage-specific repressor of transcriptional inhibitors of myelin gene expression.
  • 44.Scolding NJ, Franklin RJM, Stevens S, et al. Oligodendrocyte progenitors are present in the normal adult human CNS and in the lesions of multiple sclerosis. Brain. 1998;121:2221–2228. doi: 10.1093/brain/121.12.2221. [DOI] [PubMed] [Google Scholar]
  • 45.Wolswijk G. Chronic stage multiple sclerosis lesions contain a relatively quiescent population of oligodendrocyte precursor cells. J. Neurosci. 1998;18:601–609. doi: 10.1523/JNEUROSCI.18-02-00601.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Maeda Y, Solanky M, Menonna J, et al. Platelet-derived growth factor-alpha receptor-positive oligodendroglia are frequent in multiple sclerosis lesions. Ann. Neurol. 2001;49:776–785. doi: 10.1002/ana.1015. [DOI] [PubMed] [Google Scholar]
  • 47.Salzer JL. Polarized domains of myelinated axons. Neuron. 2003;40:297–318. doi: 10.1016/s0896-6273(03)00628-7. [DOI] [PubMed] [Google Scholar]
  • 48.Trapp BD, Peterson J, Ransohoff RM, et al. Axonal transection in the lesions of multiple sclerosis. N. Engl. J. Med. 1998;338:278–285. doi: 10.1056/NEJM199801293380502. [DOI] [PubMed] [Google Scholar]
  • 49.Kornek B, Storch MK, Weissert R, et al. Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. Am. J. Pathol. 2000;157:267–276. doi: 10.1016/S0002-9440(10)64537-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kornek B, Lassmann H. Neuropathology of multiple sclerosis – new concepts. Brain Res. Bull. 2003;61:321–326. doi: 10.1016/s0361-9230(03)00095-9. [DOI] [PubMed] [Google Scholar]
  • 51.Lappe-Siefke C, Goebbels S, Gravel M, et al. Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination. Nat. Genet. 2003;33:366–374. doi: 10.1038/ng1095. [DOI] [PubMed] [Google Scholar]
  • 52.Edgar JM, McLaughlin M, Yool D, et al. Oligodendroglial modulation of fast axonal transport in a mouse model of hereditary spastic paraplegia. J. Cell Biol. 2004;166:121–131. doi: 10.1083/jcb.200312012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Brüstle O, Jones KN, Learish RD, et al. Embryonic stem cell-derived glial precursors: a source of myelinating transplants. Science. 1999;285:754–756. doi: 10.1126/science.285.5428.754. [DOI] [PubMed] [Google Scholar]
  • 54.Utzschneider DA, Archer DR, Kocsis JD, et al. Transplantation of glial cells enhances action potential conduction of amyelinated spinal cord axons in the myelin-deficient rat. Proc. Natl. Acad. Sci. (USA) 1994;91:53–57. doi: 10.1073/pnas.91.1.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Keirstead HS, Nistor G, Bernal G, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J. Neurosci. 2005;25:4694–4705. doi: 10.1523/JNEUROSCI.0311-05.2005.. ** Demonstration of a functional effect from transplantation of human embryonic stem cells with predifferentiation into OPC indicates therapeutic potential for this approach at early stages of spinal cord injury.
  • 56.Honmou O, Felts PA, Waxman SG, Kocsis JD. Restoration of normal conduction properties in demyelinated spinal cord axons in the adult rat by transplantation of exogenous Schwann cells. J. Neurosci. 1996;16:3199–3208. doi: 10.1523/JNEUROSCI.16-10-03199.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Franklin RJ, Gilson JM, Franceschini IA, Barnett SC. Schwann cell-like myelination following transplantation of an olfactory bulb-ensheathing cell line into areas of demyelination in the adult CNS. Glia. 1996;17:217–224. doi: 10.1002/(SICI)1098-1136(199607)17:3<217::AID-GLIA4>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
  • 58.Imaizumi T, Lankford KL, Waxman SG, Greer CA, et al. Transplanted olfactory ensheathing cells remyelinate and enhance axonal conduction in the demyelinated dorsal columns of the rat spinal cord. J. Neurosci. 1998;18:6176–6185. doi: 10.1523/JNEUROSCI.18-16-06176.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Coman I, Aigrot MS, Seilhean D, Reynolds R, et al. Nodal, paranodal and juxtaparanodal axonal proteins during demyelination and remyelination in multiple sclerosis. Brain. 2006;129:3186–3195. doi: 10.1093/brain/awl144. [DOI] [PubMed] [Google Scholar]
  • 60.Lustig M, Zanazzi G, Sakurai T, et al. Nr-CAM and neurofascin interactions regulate ankyrin G and sodium channel clustering at the node of Ranvier. Curr. Biol. 2001;11:1864–1869. doi: 10.1016/s0960-9822(01)00586-3. [DOI] [PubMed] [Google Scholar]
  • 61. Howell OW, Palser A, Polito A, et al. Disruption of neurofascin localization reveals early changes preceding demyelination and remyelination in multiple sclerosis. Brain. 2006;129:3173–3185. doi: 10.1093/brain/awl290.. * Demonstration of significant alterations in oligodendrocyte Nfasc155 expression that accompany inflammation and demyelination, suggesting an ongoing disruption to the axonal-oligodendrocyte complex within newly forming as well as established lesions in multiple sclerosis.
  • 62.Charles P, Hernandez MP, Stankoff B, et al. Negative regulation of central nervous system myelination by polysialylated-neural cell adhesion molecule. Proc. Natl. Acad. Sci. (USA) 2000;97:7585–7590. doi: 10.1073/pnas.100076197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Charles P, Hernandez MP, Stankoff B, et al. Re-expression of PSA-NCAM by demyelinated axons: an inhibitor of remyelination in multiple sclerosis? Brain. 2002;125:1972–1979. doi: 10.1093/brain/awf216. [DOI] [PubMed] [Google Scholar]
  • 64. Mi S, Hu B, Hahm K, et al. LINGO-1 antagonist promotes spinal cord remyelination and axonal integrity in MOG-induced experimental autoimmune encephalomyelitis. Nat. Med. 2007;13:1228–1233. doi: 10.1038/nm1664.. ** Antagonism of LINGO-1 signaling improved axon integrity and myelination in an animal model of MS, indicating potential therapeutic benefit from approaches that attenuate LINGO-1 function.
  • 65. Lee X, Yang Z, Shao Z, et al. NGF regulates the expression of axonal LINGO-1 to inhibit oligodendrocyte differentiation and myelination. J. Neurosci. 2007;27:220–225. doi: 10.1523/JNEUROSCI.4175-06.2007.. * LINGO-1 is identified as a potent axonal inhibitor of oligodendrocyte differentiation and myelination that is regulated by nerve growth factor and its cognate receptor TrkA in a dose-dependent manner.
  • 66. Dutta R, McDonough J, Chang A, et al. Activation of the ciliary neurotrophic factor (CNTF) signaling pathway in cortical neurons of multiple sclerosis patients. Brain. 2007;130:2566–2576. doi: 10.1093/brain/awm206.. * CNTF signaling may indicate an adaptive or neuroprotective mechanisms of MS lesions that may be promising to therapeutically enhance to prevent neurodegeneration.
  • 67.Weinstock-Guttman B, Zivadinov R, Tamaño-Blanco M, et al. Immune cell BDNF secretion is associated with white matter volume in multiple sclerosis. J. Neuroimmunol. 2007;188:167–174. doi: 10.1016/j.jneuroim.2007.06.003. [DOI] [PubMed] [Google Scholar]
  • 68.Zivadinov R, Weinstock-Guttman B, Benedict R, et al. Preservation of gray matter volume in multiple sclerosis patients with the Met allele of the rs6265 (Val66Met) SNP of brain-derived neurotrophic factor. Hum. Mol. Genet. 2007;16:2659–2668. doi: 10.1093/hmg/ddm189. [DOI] [PubMed] [Google Scholar]
  • 69.Hohlfeld R. Neurotrophic talk between the nervous and immune systems; Relevance for repair strategies in multiple sclerosis? J. Neurol. Sci. 2007 Apr 23; doi: 10.1016/j.jns.2007.03.012. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 70.Sofroniew MV. Reactive astrocytes in neural repair and protection. The Neuroscientist. 2005;4:400–407. doi: 10.1177/1073858405278321. [DOI] [PubMed] [Google Scholar]
  • 71.Williams A, Piaton G, Lubetzki C. Astrocytes-friend or foes in multiple sclerosis? Glia. 2007;55:1300–1312. doi: 10.1002/glia.20546. [DOI] [PubMed] [Google Scholar]
  • 72.Pittock SJ, Lucchinetti CF. The pathology of MS: new insights and potential clinical applications. Neurologist. 2007;13:45–56. doi: 10.1097/01.nrl.0000253065.31662.37. [DOI] [PubMed] [Google Scholar]
  • 73.Blakemore WF. Regeneration and repair in multiple sclerosis: The view of experimental pathology. J. Neurol. Sci. 2007 Apr 23; doi: 10.1016/j.jns.2007.03.006. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 74. Williams A, Piaton G, Aigrot MS, et al. Semaphorin 3A and 3F: key players in myelin repair in multiple sclerosis? Brain. 2007;130:2554–2565. doi: 10.1093/brain/awm202.. *In both MS tissuer and in an experimental model of demyelination, Semaphorin 3A and 3F are detected around active demyelinating lesions, but not in chronic plaques.
  • 75.Holley JE, Gveric D, Newcombe J, et al. Astrocyte characterization in the multiple sclerosis glial scar. Neuropathol. Apll. Neurobiol. 2003;29:434–444. doi: 10.1046/j.1365-2990.2003.00491.x. [DOI] [PubMed] [Google Scholar]
  • 76.Gutowski NJ, Newcombe J, Cuzner ML. Tenascin-R and C in multiple sclerosis lesions: relevance to extracellular matrix remodelling. Neuropathol. Appl Neurobiol. 1999;25:207–214. doi: 10.1046/j.1365-2990.1999.00176.x. [DOI] [PubMed] [Google Scholar]
  • 77.Back SA, Tuohy TM, Chen H, et al. Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nat. Med. 2005;11:966–972. doi: 10.1038/nm1279. [DOI] [PubMed] [Google Scholar]
  • 78.John GR, Shankar SL, Shafit-Zagardo B, et al. Multiple sclerosis: reexpression of a developmental pathway that restricts oligodendrocyte maturation. Nat. Med. 2002;8:1115–1121. doi: 10.1038/nm781. [DOI] [PubMed] [Google Scholar]
  • 79.Fitch MT, Silver J. CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure. Exp. Neurol. 2007 May 31; doi: 10.1016/j.expneurol.2007.05.014. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ara J, See J, Mamontov P, Hahn A, Bannerman P, Pleasure D, Grinspan JB. Bone morphogenetic proteins 4, 6, and 7 are up-regulated in mouse spinal cord during experimental autoimmune encephalomyelitis. J. Neurosci. Res. 2007;86:125–135. doi: 10.1002/jnr.21462. [DOI] [PubMed] [Google Scholar]
  • 81.Cheng X, Wang Y, He Q, et al. Bone morphogenetic protein signaling and olig1/2 interact to regulate the differentiation and maturation of adult oligodendrocyte precursor cells. Stem Cells. 2007;25:3204–3214. doi: 10.1634/stemcells.2007-0284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Fuller ML, DeChant AK, Rothstein B, et al. Bone morphogenetic proteins promote gliosis in demyelinating spinal cord lesions. Ann. Neurol. 2007;62:288–300. doi: 10.1002/ana.21179. [DOI] [PubMed] [Google Scholar]
  • 83. Patrikios P, Stadelmann C, Kutzelnigg A, et al. Remyelination is extensive in a subset of multiple sclerosis patients. Brain. 2006;129:3165–3172. doi: 10.1093/brain/awl217.. ** Remyelination can occur in patients with relapsing as well as progressive MS, which may provide insight as to pathological differences in this subset of patients but also must be considered in assessing clinical trial outcomes.
  • 84. Albert M, Antel J, Brück W, Stadelmann C. Extensive cortical remyelination in patients with chronic multiple sclerosis. Brain Pathol. 2007;17:129–138. doi: 10.1111/j.1750-3639.2006.00043.x.. * In MS lesions, cortical areas may remyelinate more extensively than white matter.

RESOURCES