The contribution of oligodendrocytes and oligodendrocyte progenitor cells to central nervous system repair in multiple sclerosis: perspectives for remyelination therapeutic strategies

2018-01-05 02:54AdrianaOctavianaDulamea
中国神经再生研究(英文版) 2017年12期

Adriana Octaviana Dulamea

Department of Neurology, Fundeni Clinical Institute, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania

The contribution of oligodendrocytes and oligodendrocyte progenitor cells to central nervous system repair in multiple sclerosis: perspectives for remyelination therapeutic strategies

Adriana Octaviana Dulamea

Department of Neurology, Fundeni Clinical Institute, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania

Oligodencrocytes (OLs) are the main glial cells of the central nervous system involved in myelination of axons. In multiple sclerosis (MS), there is an imbalance between demyelination and remyelination processes, the last one performed by oligodendrocyte progenitor cells (OPCs) and OLs, resulting into a permanent demyelination, axonal damage and neuronal loss. In MS lesions, astrocytes and microglias play an important part in permeabilization of blood-brain barrier and initiation of OPCs proliferation. Migration and differentiation of OPCs are in fluenced by various factors and the process is finalized by insufficient acummulation of OLs into the MS lesion. In relation to all these processes, the author will discuss the potential targets for remyelination strategies.

multiple sclerosis; oligodencrocytes; oligodendrocyte progenitor cells; demyelination; remyelination;semaphorin; basic helix-loop-helix transcription factor oligodendrocyte transcription factor 2; leucin-rich repeat and immunoglobulin-like-domain-containing nogo receptor-interacting protein 1; canonical Notch signaling;endocrine receptors

The Role of Oligodencrocytes and Oligodendrocyte Progenitor Cells in Myelination

Oligodencrocytes (OLs) are the main glial cells of the central nervous system (CNS) involved in myelination of axons.Myelination allows saltatory conduction between nodes of Ranvier which increases both speed and energy efficiency of nerve conduction. OLs also provide trophic support to axons,lactate as energy source and have a critical role in maintenance of axonal integrity (Fünfschilling et al., 2012). However, the role of OLs in myelin sheath maintenance was uncoupled from their role in supporting axonal integrity. These results imply that OLs dysfunction alone is sufficient to cause secondary axonal degeneration and raises the possibility that OLs may be a primary cellular target in neurodegenerative disease (Tognatta and Miller, 2016).

Oligodendrocyte progenitor cells (OPCs) constitute the largest dividing population among neural cells making up on average 5% of total CNS cells and they are uniformly distributed throughout CNS (Dawson et al., 2003). They belong to the same population of progenitors that give rise to OLs during CNS development. However, a large number of OPCs do not differentiate and remain in a cyclic state during adulthood (Fernandez-Castaneda and Gaultier, 2016) until a local CNS injury occurs and triggers differentiation to OLs. During development,OPCs are generated in sequential waves from speci fic germinal sites; the first wave starts in the ventral midline of caudal regions of the neural tube as a result of inductive cues from adjacent tissues. Later in development, a second source of OPCs arises in dorsal spinal cord that generates a second wave of myelinating OLs (Cai et al., 2005) and a third wave that occurs after birth from the progenitor cells around central canal (Rowitch and Kriegstein, 2010). In more rostral regions of the CNS, cre-lox fate mapping experiments showed multiple waves of OPC generation with a ventral to dorsal progression begining at embryonic stage (Kessaris et al., 2006).

The generation of OPCs from neural progenitor cells (NPCs)continues throughout adulthood. Two primary sources of NPCs have been defined: the subventricular zone (SVZ) and the subgranular zone (SGZ) of the hippocampus. In these regions, anatomically different populations of stem cells retain the capacity to generate neurons, astrocytes and OLs (Tognatta and Miller, 2016).

In humans, most of CNS myelination occurs during the first two decades of life (Yakovlev and Lecours, 1967; Mitew et al.,2014) although there is evidence that myelination continues throughout life either to remyelinate following demyelination or brain injury or to myelinate previously unmyelinating axon(Bartzokis et al., 2012; Young et al., 2013).

Demyelination and Remyelination in Multiple Sclerosis (MS)

Persistent demyelination in MS is the result of an imbalance between dysfunction and loss of OLs that produces demyelination and the impaired/reduced generation of OLs that leads to an insufficient remyelination, evolving in parallel with axonal damage and neuronal loss.

Two patterns of OLs dysfunction can be distingushed histopatologically in MS: an immune mediated OLs dysfunction and a primary olidendrogliopathy. Studies usingin vitromodels and animal models of MS provided evidence for a direct attack to myelin and OLs of antigen speci fic cytotoxic T lymphocytes (Na et al., 2008; Lassmann, 2014) and speci fic auto-antibodies (Linington et al., 1988), T-cell mediated cytotoxicity independent from antigen recognition (Nitsch et al., 2004) as well as activation of microglia and macrophages by pro-in flammatory molecules (Felts et al., 2005). Another important player in MS lesions is the astrocyte involved in inflammation and blood-brain barrier (BBB) integrity and function. Astrocytes contribute to T cell recruitment, activation and differentiation through pro-inflammatory cytokines and chemokines production (Sørensen et al., 1999; Choi et al., 2014; Xie and Yang, 2015). They also exert,through a cell to cell contact mechanism, a direct TNF toxicity towards OPCs (Kim et al., 2011). Astrocytes secrete matrix metalloproteinases (MMPs) that increase the permeability and produce the remodeling of BBB (Williams et al., 2007). In addition, they limit the remyelination processes through interaction of NOGO with LINGO in MS plaques (Karnezis et al., 2004;Satoh et al., 2007).

Remyelination in MS occurs as a spontaneous regenerative process following demyelination (Franklin and Ffrench-Constant, 2008; Crawford et al., 2013; Aharoni, 2015) and presents greater efficiency in MS lesions appearing early in the disease course (Patrikios et al., 2006; Patani et al., 2007). In MS, the capacity of remyelination declines with age and disease progression. Remyelination in MS lesions is variable and often incomplete leading to persistent demyelination and axon degeneration(Patrikios et al., 2006; Compston and Coles, 2008; Franklin and Ffrench-Constant, 2008; Piaton et al., 2009). In general, the extent of remyelination varies from patient to patient and from a lesion to another (Zhang et al., 2016b). The remyelination is mostly restricted to the periphery of lesions, starts early in the lesion formation and is present in active lesions (Bø et al.,2013). About 10–20% of chronic lesions are completely remyelinated forming the so called shadow plaques (Patani et al.,2007). However, remyelinated regions may be more vulnerable to repeated demyelination in comparison to normally appearing white matter (NAWM) (Bramow et al., 2010) and entirely demyelinated areas are the result of repeated episodes of demyelination and incomplete remyelination (Brown et al., 2014).

Remyelination in MS was extensively studied on animal models and appears to occur in several steps. Following demyelination, factors produced by microglial cells and astrocytes activate OPCs that shift from quiescent to a regenerative fenotype that presents a different morphology and an up-regulation of several genes such as transcription factors oligodendrocyte transcription factor 2 (Olig 2), Sex determining region Y-box 2 (Sox2) and Nkx2.2 (Levine et al., 2001; Fancy et al., 2004;Talbott et al., 2005). The OPCs activation is proportional to the in flammatory reaction that succeeds demyelination and is required for succesful remyelination (Miron et al., 2011). The activated OPCs migrate to white matter lesions in response to mitogens and pro-migratory factors released by microglia and astrocytes. The migration of OPCs appears to be regulated by chemo-attractant factors such as platelet derived growth factor (PDGF) and semaphorin 3F (Sema 3F), chemo-reppelents netrin-1, semaphorin 3A (Sema 3A), ephrins and stop-signals chemokine (CXCL)1 and tenascin C (Dubois-Dalcq and Murray, 2000; Kakinuma et al., 2004; Sobel, 2005; Williams et al.,2007; Kerstetter et al., 2009; Miron et al., 2011; Bin et al., 2013;Boyd et al., 2013).

To populate demyelinated areas, the recruited OPCs start to differentiate into remyelinating OLs (Franklin and Ffrench-Constant, 2008; Bradl and Lassmann, 2010). The differentiation of OPCs is promoted by insulin-like growth factor(IGF-1), ciliary neurotrophic factor (CNTF) and thyroid hormone (Zhang et al., 2015, 2016a) and requires the function of Olig1, Olig2, Nkx2.6, Myt1 and sex determining region Y box(SOX)-10 (Nunes et al., 2003; Fancy et al., 2004; Nicolay et al.,2007). Then OLs need to establish a contact with the axon to be remyelinated, before generating the myelin protein membrane.Axon-glia interaction and myelin membrane tra ficking are essential for remyelination. Src-family kinase Fyn plays a central role in axonal signal integration by OLs (White and Krämer-Albers, 2014). The development of myelin sheath in remyelination follows a similar pattern with developmental myelination,although the rate of OPCs migration is slower and the myelin sheaths in remyelinated areas are thinner and shorter but suffi-cient to ensure full functional recovery of the axons (Bradl and Lassmann, 2010; Fancy et al., 2010; Crawford et al., 2013).

Recent animal models studies, using genetic fate mapping techniques, implicate OPCs as the cells responsible for remyelination and not the mature previously myelinating OLs (Tripathi et al., 2010; Zawadzka et al., 2010).

Causes of Remyelination Failure in MS

The causes of remyelination failure in MS appear to be related to the depletion of OPCs as well as the impaired migration and differentiation of OPCs (Figure 1).

Thus, OPCs were identified within active MS lesions but their number and capacity to differentiate decreases with disease duration and OPCs were more vulnerable to injury mediators than OLs (Chang et al., 2002). However, depletion of OPCs is not the only cause of remyelination failure since studies of autopsy from MS patients showed the presence of OPCs and immature OLs contacting the axons but failing to myelinate (Câmara and ffrench-Constant, 2007). Also, analysis of focal demyelinated lesions showed that areas of succesive demyelination-remyelination as well as first time remyelinated areas presented no evidence of OPCs depletion and, in chronic MS lesions, OPCs are present but fail to remyelinate (Kuhlmann et al., 2008; Hartley et al., 2014). In chronic lesions remyelination impairment is due to de ficient OPCs recruitment phase which involves proliferation, migration and repopulation of lesions(Figure 2). Activated microglia and astrocytes in the setting of demyelination are the source of mitogens/growth factors such as fibroblast growth factor 2 (FGF-2) and PDGF-2A that induce rapid proliferative responses in OPCs (Armstrong et al., 2002; Woodruff et al., 2004). This OPCs response is regulated by p27Kip1 (Crockett et al., 2005) and Cdk (Caillava and Baron-Van Evercooren, 2012; Tognatta and Miller, 2016). In chronic MS lesions the changes in the extracellular matrix and the formation of the astroglial scar (Franklin and Ffrench-Constant, 2008) contribute to reduction of proliferation, migration and accumulation of OPCs in the lesion (Franklin, 2002).

Several studies identi fied inhibitors of OPCs migration during remyelination but their action is not well understood (Piaton et al., 2011). One of the factors that modulate OPCs migration are group 3 semaphorins, Sema 3A is an inhibitory in contrast to Sema 3F which is an attractive migratory signal for OPCs.Active MS lesions contain higher mRNA expression of chemoattractant Sema 3F than chemoreppelent Sema 3A (Williams et al., 2007). Another study analyzed a series of MS lesions in postmortem tissue and found a correlation between a lower number of OPCs, chronic active lesion type and a higher expression of chemoreppelent Sema 3A (Boyd et al., 2013). In contrast, a low expression of Sema 3A and a higher expression of chemoattractant Sema 3F correlated with active lesions and more variable,but generally higher numbers of OPCs.

Another factor that in fluences OPCs migration and remyelination is chemoreppelent netrin-1. The role of netrin-1 as a repellent for migrating OPCs during development (Sugimoto et al., 2001; Jarjour et al., 2003; Bin et al., 2013; Tepavčević et al., 2014) and its expression by neurons and glia in the mature CNS (Manitt et al., 2001) suggest that it might also in fluence OPCs migration and remyelination in MS. Full length and fragmented netrin-1 were found in adult human white matter as well as in demyelinated MS lesions, where they exert inhibitory in fluences on OPCs migration (Manitt et al., 2006; Löw et al., 2008; Bin et al., 2013).

Chemokines exert various effects on migration of OPCs and OLs and differentiation of OPCs throughout the process of remyelination. OLs express at least four chemokine receptors: CXCR1,CXCR2, CXCR3 and CXCR4 (Nguyen and Stangel, 2001; Omari et al., 2005). Histological studies suggest that chemokine CXCL1 is upregulated around the peripheral areas of demyelination.Also, localized inhibition of CXCR2 signaling reduces lesion size and enhances remyelination (Kerstetter et al., 2009). CXCL12 was signi ficantly upregulated within activated astrocytes and microglia during demyelination, as were numbers of CXCR4+NG2+OPCs. Loss of CXCR4 signalingviaeither pharmacological blockade orin vivoRNA silencing led to decreased OPCs maturation and failure to remyelinate (Patel et al., 2010).

The differentiation of OPCs is regulated by a complex interplay of intrinsic, extrinsic and epigenetic factors during development (Rowitch and Kriegstein, 2010), some of them being also involved in remyelination in MS.

Among intrinsic factors, the basic helix-loop-helix (bHLH)transcription factor Olig2 has a critical role in OLs determination (Rowitch, 2004). Wegener et al. (2015) demonstrate that Olig2 displays a differential expression pattern in MS lesions that correlates to lesion activity. Olig2 was predominantly detected in NOGO-A+(now known as RTN4-A) maturing OLs,which prevale in active lesion borders, rather than chronic silent and shadow plaques.

Figure 1 Causes of remyelination failure in MS.

Figure 2 Factors in fluencing remyelination in MS lesions.

Leucin-rich repeat and immunoglobulin-like-domain-containing nogo receptor-interacting protein 1 (LINGO-1) is a potent negative regulator of neuron and OL survival, neurite extension,axon regeneration, OL differentiation, axonal myelination and functional recovery (Yin and Hu, 2014). In experimental autoimmune encephalomyelitis (EAE) models of MS, LINGO-1 knockout mice exhibit enhanced myelin sheath formation and recovery (Mi et al., 2007) and treatment with LINGO-1 antagonist result in increased OPCs differentiation and remyelination(Zhang et al., 2015, 2016a) suggesting that blocking LINGO-1 may be a useful therapeutic approach (Rudick et al., 2008). In a phase I clinical trial, anti-LINGO-1 monoclonal antibody(BIIB033) showed safety and tolerability in MS patients (Tran et al., 2014). Further more, two phase two clinical studies have been started. A phase II placebo-controlled clinical trial (RENEW)in 82 patients with optic neuritis who received six intravenous infusions of opicimumab or placebo every four weeks, followed up for 32 weeks, showed that opicimumab was no better than placebo for improving visual function but statistically signi ficant improved the visual evoked potentials (Cadavid et al., 2017). The second phase II clinical trial (SYNERGY) including 418 relapsing remitting or secondary progressive MS patients showed that opicimumab had satisfactory tolerability, however did not lead to an improvement in disability or a slow down in disability progression but there were indications of a clinical effect that will be further studied (McCroskery et al., 2017).

Canonical Notch signaling, which occurs through ligands such as Jagged, inhibits OPCs differentiation during development(Genoud et al., 2002), however its role in CNS remyelination is still debated. Hammond et al. (2014) recently demonstrated that OPC differentiation following lysolecithin demyelination is inhibited by Jagged1-expressing astrocytes, which directly bind to Notch1 on OPCs. Reactive astrocytes express Jagged1 in MS plaques (John et al., 2002) and this expression appears to be regulated by the secreted protein endothelin-1 (ET-1) (Hammond et al., 2014), which inhibits OL differentiation during development (Chamberlain et al., 2016).

Wnt proteins are secreted ligands that play numerous roles in regulating development, including OLs genesis (Ortega et al., 2013). However the contribution of Wnt pathway in remyelination process is controversial and targeting Wnt pathway as potential remyelinating therapy must be approached with caution (Xie et al., 2014; Guo et al., 2015).

Nuclear retinoid X receptor (RXR) pathway plays an important role in cell proliferation and development (Mark et al., 2009). RXR couple with retinoic acid receptor, vitamin D receptor (VDR), thyroid receptor and peroxisome proliferator-activated receptor to induce gene transcription (Rastinejad,2001). RXR-VDR signaling induces OPCs differentiation and VDR agonist vitamin D enhances OPCs differentiation (de la Fuente et al., 2015). Several studies showed expression of VDR in oligodendroglial lineage cells in MS, revealing a potential role of vitamin D in remyelination (Ballanger et al., 2010; de la Fuente et al., 2015).

Other potential remyelination targets are endocrine receptors. MS shows a female-to-male gender prevalence and disturbances in sex steroid production (Kipp et al., 2012). Estrogen and progesterone operate in reducing central and brain-intrinsic immune responses and regulating local growth factors supply, OL and astrocyte function (Kipp et al., 2012). Several studies showed that low estrogen states favor exacerbations of MS in women (Christianson et al., 2015; Triantafyllou et al.,2016) and levels of progesterone and testosterone metabolites are decreased in cerebrospinal fluid of MS patients (Caruso et al., 2014). Endocrine targets have been investigated in preclinical models of MS as potential modulators of myelination.Numerous preclinical studies demonstrated that 17-β estradiol, estriol and other estrogen receptor (ER) ligand treatments have protective effects on susceptibility to EAE (Crawford et al., 2010), thyroid hormones promote myelin repair (Harsan et al., 2008; Calza et al., 2010; D’Intino et al., 2011), progesterone decreases demyelination, disease severity and neurological deficits (Schumacher et al., 2012) and testosterone enhances remyelination through neural androgen receptor (Hussain et al., 2013). However, clinical trials using estrogen therapy in MS patients showed an increased risk of breast and uterine cancer,heart disease and stroke (Prentice et al., 2009), the majority of these effects being mediated through ER α (Caringella et al., 2011). As a result, ERβ became interesting as a target for neuroprotective therapy (Planey et al., 2014). Other clinical trials showed that testosterone treatment in MS patients was correlated to reduced inflammation and improved cognition(Sicotte et al., 2007; Gold et al., 2008).

Conclusion

OLs and OPCs are essential cells for myelination of the CNS.The process of myelination starts during development and continues throughout life under the control of a complex genetic mechanism. In MS, the immune process induces a pathological cascade that injures OLs and OPCs generating demyelination and impaired remyelination. The identi fication of key molecules and pathways controlling the migration and differentiation of OPCs and myelination has provided clues for potential targets of drug candidates in order to develop effi-cient remyelination strategies for MS.

Author contributions:AOD wrote, edited, and revised the contents of this paper and created the figures.

Con flicts of interest:None declared.

Plagiarism check:Checked twice by iThenticate.

Peer review:Externally peer reviewed.

Open access statement:This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 3.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under identical terms.

Aharoni R (2015) Remyelination in multiple sclerosis: realizing a long-standing challenge. Expert Rev Neurother 15:1369-1372.

Armstrong RC, Le TQ, Frost EE, Borke RC, Vana AC (2002) Absence of fibroblast growth factor 2 promotes oligodendroglial repopulation of demyelinated white matter. J Neurosci 22:8574-8585.

Bø L, Esiri M, Evangelou N, Kuhlmann T (2013) Demyelination and Remyelination in Multiple Sclerosis. In: Myelin Repair and Neuroprotection in Multiple Sclerosis (Duncan ID, Franklin RJM, eds), pp 23-45. Boston, MA:Springer US.

Ballanger F, Nguyen JM, Khammari A, Dréno B (2010) Evolution of clinical and molecular responses to bexarotene treatment in cutaneous T-cell lymphoma. Dermatology 220:370-375.

Bartzokis G, Lu PH, Heydari P, Couvrette A, Lee GJ, Kalashyan G, Freeman F,Grinstead JW, Villablanca P, Finn JP, Mintz J, Alger JR, Altshuler LL (2012)Multimodal magnetic resonance imaging assessment of white matter aging trajectories over the lifespan of healthy individuals. Biol Psychiatry 72:1026-1034.

Bin JM, Rajasekharan S, Kuhlmann T, Hanes I, Marcal N, Han D, Rodrigues SP, Leong SY, Newcombe J, Antel JP, Kennedy TE (2013) Full-length and fragmented netrin-1 in multiple sclerosis plaques are inhibitors of oligodendrocyte precursor cell migration. Am J Pathol 183:673-680.

Boyd A, Zhang H, Williams A (2013) Insufficient OPC migration into demyelinated lesions is a cause of poor remyelination in MS and mouse models.Acta Neuropathol 125:841-859.

Bradl M, Lassmann H (2010) Oligodendrocytes: biology and pathology. Acta Neuropathol 119:37-53.

Bramow S, Frischer JM, Lassmann H, Koch-Henriksen N, Lucchinetti CF,Sorensen PS, Laursen H (2010) Demyelination versus remyelination in progressive multiple sclerosis. Brain 133:2983-2998.

Brown RA, Narayanan S, Arnold DL (2014) Imaging of repeated episodes of demyelination and remyelination in multiple sclerosis. Neuroimage Clin 6:20-25.

Câmara J, ffrench-Constant C (2007) Lessons from oligodendrocyte biology on promoting repair in multiple sclerosis. J Neurol 254:I15-I22.

Cadavid D, Balcer L, Galetta S, Aktas O, Ziemssen T, Vanopdenbosch L,Frederiksen J, Skeen M, Jaffe GJ, Butzkueven H, Ziemssen F, Massacesi L,Chai Y, Xu L, Freeman S; RENEW Study Investigators (2017) Safety and efficacy of opicinumab in acute optic neuritis (RENEW): a randomised,placebo-controlled, phase 2 trial. Lancet Neurol 16:189-199.

Cai J, Qi Y, Hu X, Tan M, Liu Z, Zhang J, Li Q, Sander M, Qiu M (2005)Generation of oligodendrocyte precursor cells from mouse dorsal spinal cord independent of Nkx6 regulation and Shh signaling. Neuron 45:41-53.

Caillava C, Baron-Van Evercooren A (2012) Differential requirement of cyclin-dependent kinase 2 for oligodendrocyte progenitor cell proliferation and differentiation. Cell division 7:14.

Calza L, Fernandez M, Giardino L (2010) Cellular approaches to central nervous system remyelination stimulation: thyroid hormone to promote myelin repair via endogenous stem and precursor cells. J Mol Endocrinol 44:13-23.

Caringella AM, Di Naro E, Loverro G (2011) Clinical function of estrogen receptors in endometrial cancer. Minerva Ginecol 63:495-504.

Caruso D, Melis M, Fenu G, Giatti S, Romano S, Grimoldi M, Crippa D,Marrosu MG, Cavaletti G, Melcangi RC (2014) Neuroactive steroid levels in plasma and cerebrospinal fluid of male multiple sclerosis patients. J Neurochem 130:591-597.

Chamberlain KA, Nanescu SE, Psachoulia K, Huang JK (2016) Oligodendrocyte regeneration: Its signi ficance in myelin replacement and neuroprotection in multiple sclerosis. Neuropharmacology 110:633-643.

Chang A, Tourtellotte WW, Rudick R, Trapp BD (2002) Premyelinating oligodendrocytes in chronic lesions of multiple sclerosis. N Engl J Med 346:165-173.

Choi SS, Lee HJ, Lim I, Satoh J, Kim SU (2014) Human astrocytes: secretome pro files of cytokines and chemokines. PLoS One 9:e92325.

Christianson MS, Mensah VA, Shen W (2015) Multiple sclerosis at menopause: Potential neuroprotective effects of estrogen. Maturitas 80:133-139.

Compston A, Coles A (2008) Multiple sclerosis. Lancet 372:1502-1517.

Crawford AH, Chambers C, Franklin RJ (2013) Remyelination: the true regeneration of the central nervous system. J Comp Pathol 149:242-254.

Crawford DK, Mangiardi M, Song B, Patel R, Du S, Sofroniew MV, Voskuhl RR, Tiwari-Woodruff SK (2010) Oestrogen receptor beta ligand: a novel treatment to enhance endogenous functional remyelination. Brain 133:2999-3016.

Crockett DP, Burshteyn M, Garcia C, Muggironi M, Casaccia-Bonnefil P(2005) Number of oligodendrocyte progenitors recruited to the lesioned spinal cord is modulated by the levels of the cell cycle regulatory protein p27Kip-1. Glia 49:301-308.

D’Intino G, Lorenzini L, Fernandez M, Taglioni A, Perretta G, Del Vecchio G, Villoslada P, Giardino L, Calza L (2011) Triiodothyronine administration ameliorates the demyelination/remyelination ratio in a non-human primate model of multiple sclerosis by correcting tissue hypothyroidism. J Neuroendocrinol 23:778-790.

Dawson MR, Polito A, Levine JM, Reynolds R (2003) NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci 24:476-488.

de la Fuente AG, Errea O, van Wijngaarden P, Gonzalez GA, Kerninon C,Jarjour AA, Lewis HJ, Jones CA, Nait-Oumesmar B, Zhao C, Huang JK,ffrench-Constant C, Franklin RJ (2015) Vitamin D receptor-retinoid X receptor heterodimer signaling regulates oligodendrocyte progenitor cell differentiation. J Cell Biol 211:975-985.

Dubois-Dalcq M, Murray K (2000) Why are growth factors important in oligodendrocyte physiology? Pathol Biol (Paris) 48:80-86.

Fünfschilling U, Supplie LM, Mahad D, Boretius S, Saab AS, Edgar J, Brinkmann BG, Kassmann CM, Tzvetanova ID, Mobius W, Diaz F, Meijer D,Suter U, Hamprecht B, Sereda MW, Moraes CT, Frahm J, Goebbels S,Nave K-A (2012) Glycolytic oligodendrocytes maintain myelin and longterm axonal integrity. Nature 485:517-521.

Fancy SP, Zhao C, Franklin RJ (2004) Increased expression of Nkx2.2 and Olig2 identifies reactive oligodendrocyte progenitor cells responding to demyelination in the adult CNS. Mol Cell Neurosci 27:247-254.

Fancy SP, Kotter MR, Harrington EP, Huang JK, Zhao C, Rowitch DH,Franklin RJ (2010) Overcoming remyelination failure in multiple sclerosis and other myelin disorders. Exp Neurol 225:18-23.

Felts PA, Woolston AM, Fernando HB, Asquith S, Gregson NA, Mizzi OJ,Smith KJ (2005) In flammation and primary demyelination induced by the intraspinal injection of lipopolysaccharide. Brain 128:1649-1666.

Fernandez-Castaneda A, Gaultier A (2016) Adult oligodendrocyte progenitor cells - Multifaceted regulators of the CNS in health and disease. Brain Behav Immun 57:1-7.

Franklin RJ (2002) Why does remyelination fail in multiple sclerosis? Nat Rev Neurosci 3:705-714.

Franklin RJ, Ffrench-Constant C (2008) Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci 9:839-855.

Genoud S, Lappe-Siefke C, Goebbels S, Radtke F, Aguet M, Scherer SS, Suter U, Nave KA, Mantei N (2002) Notch1 control of oligodendrocyte differentiation in the spinal cord. J Cell Biol 158:709-718.

Gold SM, Chalifoux S, Giesser BS, Voskuhl RR (2008) Immune modulation and increased neurotrophic factor production in multiple sclerosis patients treated with testosterone. J Neuroin flammation 5:32.

Guo F, Lang J, Sohn J, Hammond E, Chang M, Pleasure D (2015) Canonical Wnt signaling in the oligodendroglial lineage--puzzles remain. Glia 63:1671-1693.

Hammond TR, Gadea A, Dupree J, Kerninon C, Nait-Oumesmar B, Aguirre A, Gallo V (2014) Astrocyte-derived endothelin-1 inhibits remyelination through notch activation. Neuron 81:588-602.

Harsan LA, Steibel J, Zaremba A, Agin A, Sapin R, Poulet P, Guignard B,Parizel N, Grucker D, Boehm N, Miller RH, Ghandour MS (2008) Recovery from chronic demyelination by thyroid hormone therapy: myelinogenesis induction and assessment by diffusion tensor magnetic resonance imaging. J Neurosci 28:14189-14201.

Hartley MD, Altowaijri G, Bourdette D (2014) Remyelination and multiple sclerosis: therapeutic approaches and challenges. Curr Neurol Neurosci Rep 14:485.

Hussain R, Ghoumari AM, Bielecki B, Steibel J, Boehm N, Liere P, Macklin WB, Kumar N, Habert R, Mhaouty-Kodja S, Tronche F, Sitruk-Ware R, Schumacher M, Ghandour MS (2013) The neural androgen receptor:a therapeutic target for myelin repair in chronic demyelination. Brain 136:132-146.

Jarjour AA, Manitt C, Moore SW, Thompson KM, Yuh SJ, Kennedy TE (2003)Netrin-1 is a chemorepellent for oligodendrocyte precursor cells in the embryonic spinal cord. J Neurosci 23:3735-3744.

John GR, Shankar SL, Sha fit-Zagardo B, Massimi A, Lee SC, Raine CS, Brosnan CF (2002) Multiple sclerosis: re-expression of a developmental pathway that restricts oligodendrocyte maturation. Nat Med 8:1115-1121.

Kakinuma Y, Saito F, Osawa S, Miura M (2004) A mechanism of impaired mobility of oligodendrocyte progenitor cells by tenascin C through modification of wnt signaling. FEBS Lett 568:60-64.

Karnezis T, Mandemakers W, McQualter JL, Zheng B, Ho PP, Jordan KA,Murray BM, Barres B, Tessier-Lavigne M, Bernard CC (2004) The neurite outgrowth inhibitor Nogo A is involved in autoimmune-mediated demyelination. Nat Neurosci 7:736-744.

Kerstetter AE, Padovani-Claudio DA, Bai L, Miller RH (2009) Inhibition of CXCR2 signaling promotes recovery in models of multiple sclerosis. Exp Neurol 220:44-56.

Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson WD(2006) Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci 9:173-179.

Kim S, Steelman AJ, Koito H, Li J (2011) Astrocytes promote TNF-mediated toxicity to oligodendrocyte precursors. J Neurochem 116:53-66.

Kipp M, Amor S, Krauth R, Beyer C (2012) Multiple sclerosis: neuroprotective alliance of estrogen-progesterone and gender. Front Neuroendocrinol 33:1-16.

Kuhlmann T, Miron V, Cui Q, Wegner C, Antel J, Bruck W (2008) Differentiation block of oligodendroglial progenitor cells as a cause for remyelination failure in chronic multiple sclerosis. Brain 131:1749-1758.

Löw K, Culbertson M, Bradke F, Tessier-Lavigne M, Tuszynski MH (2008)Netrin-1 is a novel myelin-associated inhibitor to axon growth. J Neurosci 28:1099-1108.

Lassmann H (2014) Multiple sclerosis: lessons from molecular neuropathology. Exp Neurol 262 Pt A:2-7.

Levine JM, Reynolds R, Fawcett JW (2001) The oligodendrocyte precursor cell in health and disease. Trends Neurosci 24:39-47.

Linington C, Bradl M, Lassmann H, Brunner C, Vass K (1988) Augmentation of demyelination in rat acute allergic encephalomyelitis by circulating mouse monoclonal antibodies directed against a myelin/oligodendrocyte glycoprotein. Am J Pathol 130:443-454.

Manitt C, Wang D, Kennedy TE, Howland DR (2006) Positioned to inhibit:netrin-1 and netrin receptor expression after spinal cord injury. J Neurosci Res 84:1808-1820.

Manitt C, Colicos MA, Thompson KM, Rousselle E, Peterson AC, Kennedy TE (2001) Widespread expression of netrin-1 by neurons and oligodendrocytes in the adult mammalian spinal cord. J Neurosci 21:3911-3922.

Mark M, Ghyselinck NB, Chambon P (2009) Function of retinoic acid receptors during embryonic development. Nucl Recept Signal 7:e002.

McCroskery P, Selmaj K, Fernandez O, Grimaldi LME, Silber E, Pardo G,Freedman SM, Zhang Y, Xu L, Cadavid D, Mellion M (2017) Safety and tolerability of opicinumab in relapsing multiple sclerosis: the phase 2b SYNERGY trial (P5.369). Neurology 88:P5.369.

Mi S, Hu B, Hahm K, Luo Y, Kam Hui ES, Yuan Q, Wong WM, Wang L, Su H, Chu TH, Guo J, Zhang W, So KF, Pepinsky B, Shao Z, Graff C, Garber E, Jung V, Wu EX, Wu W (2007) LINGO-1 antagonist promotes spinal cord remyelination and axonal integrity in MOG-induced experimental autoimmune encephalomyelitis. Nat Med 13:1228-1233.

Miron VE, Kuhlmann T, Antel JP (2011) Cells of the oligodendroglial lineage,myelination, and remyelination. Biochim Biophys Acta 1812:184-193.

Mitew S, Hay CM, Peckham H, Xiao J, Koenning M, Emery B (2014) Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 276:29-47.

Na SY, Cao Y, Toben C, Nitschke L, Stadelmann C, Gold R, Schimpl A, Hunig T (2008) Naive CD8 T-cells initiate spontaneous autoimmunity to a sequestered model antigen of the central nervous system. Brain 131:2353-2365.

Nguyen D, Stangel M (2001) Expression of the chemokine receptors CXCR1 and CXCR2 in rat oligodendroglial cells. Brain Res Dev Brain Res 128:77-81.Nicolay DJ, Doucette JR, Nazarali AJ (2007) Transcriptional control of oligodendrogenesis. Glia 55:1287-1299.

Nitsch R, Pohl EE, Smorodchenko A, Infante-Duarte C, Aktas O, Zipp F(2004) Direct impact of T cells on neurons revealed by two-photon microscopy in living brain tissue. J Neurosci 24:2458-2464.

Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G, 2nd, Jiang L,Kang J, Nedergaard M, Goldman SA (2003) Identi fication and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain. Nat Med 9:439-447.

Omari KM, John GR, Sealfon SC, Raine CS (2005) CXC chemokine receptors on human oligodendrocytes: implications for multiple sclerosis. Brain 128:1003-1015.

Ortega F, Gascon S, Masserdotti G, Deshpande A, Simon C, Fischer J, Dimou L, Chichung Lie D, Schroeder T, Berninger B (2013) Oligodendrogliogenic and neurogenic adult subependymal zone neural stem cells constitute distinct lineages and exhibit differential responsiveness to Wnt signalling.Nat Cell Biol 15:602-613.

Patani R, Balaratnam M, Vora A, Reynolds R (2007) Remyelination can be extensive in multiple sclerosis despite a long disease course. Neuropathol Appl Neurobiol 33:277-287.

Patel JR, McCandless EE, Dorsey D, Klein RS (2010) CXCR4 promotes differentiation of oligodendrocyte progenitors and remyelination. Proc Natl Acad Sci U S A 107:11062-11067.

Patrikios P, Stadelmann C, Kutzelnigg A, Rauschka H, Schmidbauer M,Laursen H, Sorensen PS, Bruck W, Lucchinetti C, Lassmann H (2006)Remyelination is extensive in a subset of multiple sclerosis patients. Brain 129:3165-3172.

Piaton G, Williams A, Seilhean D, Lubetzki C (2009) Remyelination in multiple sclerosis. Prog Brain Res 175:453-464.

Piaton G, Aigrot MS, Williams A, Moyon S, Tepavcevic V, Moutkine I, Gras J,Matho KS, Schmitt A, Soellner H, Huber AB, Ravassard P, Lubetzki C (2011)Class 3 semaphorins in fluence oligodendrocyte precursor recruitment and remyelination in adult central nervous system. Brain 134:1156-1167.

Planey SL, Kumar R, Arnott JA (2014) Estrogen receptors (ERalpha versus ERbeta): friends or foes in human biology? J Recept Signal Transduct Res 34:1-5.

Prentice RL, Huang Y, Hinds DA, Peters U, Pettinger M, Cox DR, Beilharz E, Chlebowski RT, Rossouw JE, Caan B, Ballinger DG (2009) Variation in the FGFR2 gene and the effects of postmenopausal hormone therapy on invasive breast cancer. Cancer Epidemiol Biomarkers Prev 18:3079-3085.

Rastinejad F (2001) Retinoid X receptor and its partners in the nuclear receptor family. Curr Opin Struct Biol 11:33-38.

Rowitch DH (2004) Glial speci fication in the vertebrate neural tube. Nat Rev Neurosci 5:409-419.

Rowitch DH, Kriegstein AR (2010) Developmental genetics of vertebrate glial-cell speci fication. Nature 468:214-222.

Rudick RA, Mi S, Sandrock AW, Jr. (2008) LINGO-1 antagonists as therapy for multiple sclerosis: in vitro and in vivo evidence. Expert Opin Biol Ther 8:1561-1570.

Sørensen TL, Tani M, Jensen J, Pierce V, Lucchinetti C, Folcik VA, Qin S,Rottman J, Sellebjerg F, Strieter RM, Frederiksen JL, Ransohoff RM (1999)Expression of speci fic chemokines and chemokine receptors in the central nervous system of multiple sclerosis patients. J Clin Invest 103:807-815.

Satoh J, Tabunoki H, Yamamura T, Arima K, Konno H (2007) TROY and LINGO-1 expression in astrocytes and macrophages/microglia in multiple sclerosis lesions. Neuropathol Appl Neurobiol 33:99-107.

Schumacher M, Hussain R, Gago N, Oudinet JP, Mattern C, Ghoumari AM(2012) Progesterone synthesis in the nervous system: implications for myelination and myelin repair. Front Neurosci 6:10.

Sicotte NL, Giesser BS, Tandon V, Klutch R, Steiner B, Drain AE, Shattuck DW, Hull L, Wang HJ, Elashoff RM, Swerdloff RS, Voskuhl RR (2007)Testosterone treatment in multiple sclerosis: a pilot study. Arch Neurol 64:683-688.

Sobel RA (2005) Ephrin A receptors and ligands in lesions and normal-appearing white matter in multiple sclerosis. Brain Pathol 15:35-45.

Sugimoto Y, Taniguchi M, Yagi T, Akagi Y, Nojyo Y, Tamamaki N (2001)Guidance of glial precursor cell migration by secreted cues in the developing optic nerve. Development 128:3321-3330.

Talbott JF, Loy DN, Liu Y, Qiu MS, Bunge MB, Rao MS, Whittemore SR(2005) Endogenous Nkx2.2+/Olig2+ oligodendrocyte precursor cells fail to remyelinate the demyelinated adult rat spinal cord in the absence of astrocytes. Exp Neurol 192:11-24.

Tepavčević V, Kerninon C, Aigrot MS, Meppiel E, Mozafari S, Arnould-Laurent R, Ravassard P, Kennedy TE, Nait-Oumesmar B, Lubetzki C (2014)Early netrin-1 expression impairs central nervous system remyelination.Ann Neurol 76:252-268.

Tognatta R, Miller RH (2016) Contribution of the oligodendrocyte lineage to CNS repair and neurodegenerative pathologies. Neuropharmacology 110:539-547.

Tran JQ, Rana J, Barkhof F, Melamed I, Gevorkyan H, Wattjes MP, de Jong R,Brosofsky K, Ray S, Xu L, Zhao J, Parr E, Cadavid D (2014) Randomized phase I trials of the safety/tolerability of anti-LINGO-1 monoclonal antibody BIIB033. Neurol Neuroimmunol Neuroin flamm 1:e18.

Triantafyllou N, Thoda P, Armeni E, Rizos D, Kaparos G, Augoulea A, Alexandrou A, Creatsa M, Tsivgoulis G, Artemiades A, Panoulis C, Lambrinoudaki I (2016) Association of sex hormones and glucose metabolism with the severity of multiple sclerosis. Int J Neurosci 126:797-804.

Tripathi RB, Rivers LE, Young KM, Jamen F, Richardson WD (2010) NG2 glia generate new oligodendrocytes but few astrocytes in a murine experimental autoimmune encephalomyelitis model of demyelinating disease. J Neurosci 30:16383-16390.

Wegener A, Deboux C, Bachelin C, Frah M, Kerninon C, Seilhean D, Weider M, Wegner M, Nait-Oumesmar B (2015) Gain of Olig2 function in oligodendrocyte progenitors promotes remyelination. Brain 138:120-135.

White R, Krämer-Albers EM (2014) Axon-glia interaction and membrane traffic in myelin formation. Front Cell Neurosci 7:284.

Williams A, Piaton G, Aigrot MS, Belhadi A, Theaudin M, Petermann F,Thomas JL, Zalc B, Lubetzki C (2007) Semaphorin 3A and 3F: key players in myelin repair in multiple sclerosis? Brain 130:2554-2565.

Woodruff RH, Fruttiger M, Richardson WD, Franklin RJ (2004) Platelet-derived growth factor regulates oligodendrocyte progenitor numbers in adult CNS and their response following CNS demyelination. Mol Cell Neurosci 25:252-262.

Xie C, Li Z, Zhang GX, Guan Y (2014) Wnt signaling in remyelination in multiple sclerosis: friend or foe? Mol Neurobiol 49:1117-1125.

Xie L, Yang SH (2015) Interaction of astrocytes and T cells in physiological and pathological conditions. Brain Res 1623:63-73.

Yakovlev P, Lecours A (1967) The myelogenetic cycles of regional maturation of the brain. In: Regional Development of the Brain in Early Life (Minkowski A, ed), pp 3-70. Blackwell: Oxford, UK.

Yin W, Hu B (2014) Knockdown of Lingo1b protein promotes myelination and oligodendrocyte differentiation in zebra fish. Exp Neurol 251:72-83.

Young KM, Psachoulia K, Tripathi RB, Dunn SJ, Cossell L, Attwell D, Tohyama K, Richardson WD (2013) Oligodendrocyte dynamics in the healthy adult CNS: evidence for myelin remodeling. Neuron 77:873-885.

Zawadzka M, Rivers LE, Fancy SP, Zhao C, Tripathi R, Jamen F, Young K,Goncharevich A, Pohl H, Rizzi M, Rowitch DH, Kessaris N, Suter U, Richardson WD, Franklin RJ (2010) CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell 6:578-590.

Zhang M, Ma Z, Qin H, Yao Z (2016a) Thyroid hormone potentially bene fits multiple sclerosis via facilitating remyelination. Mol Neurobiol 53:4406-4416.

Zhang Y, Zhang YP, Pepinsky B, Huang G, Shields LB, Shields CB, Mi S (2015)Inhibition of LINGO-1 promotes functional recovery after experimental spinal cord demyelination. Exp Neurol 266:68-73.

Zhang Y, Jonkman L, Klauser A, Barkhof F, Yong VW, Metz LM, Geurts JJ(2016b) Multi-scale MRI spectrum detects differences in myelin integrity between MS lesion types. Mult Scler 22:1569-1577.

How to cite this article:Dulamea AO (2017) The contribution of oligodendrocytes and oligodendrocyte progenitor cells to central nervous system repair in multiple sclerosis: perspectives for remyelination therapeutic strategies. Neural Regen Res 12(12):1939-1944.

*Correspondence to:Adriana Octaviana Dulamea,M.D.,adrianadulamea@gmail.com.

orcid:0000-0002-5056-0191(Adriana Octaviana Dulamea)

10.4103/1673-5374.221146

2017-11-22