378
Views
3
CrossRef citations to date
0
Altmetric
Review

Regenerative replacement of neural cells for treatment of spinal cord injury

, , & ORCID Icon
Pages 1411-1427 | Received 08 Oct 2020, Accepted 06 Apr 2021, Published online: 26 Apr 2021

References

  • Choo AM, Liu J, Lam CK, et al. Contusion, dislocation, and distraction: primary hemorrhage and membrane permeability in distinct mechanisms of spinal cord injury. J Neurosurg Spine. 2007;6(3):255–266.
  • Rowland JW, Hawryluk GWJ, Kwon B, et al. Current status of acute spinal cord injury pathophysiology and emerging therapies: promise on the horizon. Neurosurg Focus. 2008;25:E2.
  • Sekhon LH, Fehlings MG. Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine (Phila Pa 1976). 2001;26(Supplement):S2–12.
  • Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic spinal cord injury: an overview of pathophysiology, models and acute injury mechanisms. Front Neurol. 2019;10:282.
  • Xu W, Chi L, Xu R, et al. Increased production of reactive oxygen species contributes to motor neuron death in a compression mouse model of spinal cord injury. Spinal Cord. 2005;43(4):204–213.
  • Hassannejad Z, Zadegan SA, Shakouri-Motlagh A, et al. The fate of neurons after traumatic spinal cord injury in rats: a systematic review. Iran J Basic Med Sci. 2018;21:546–557.
  • Arrázola MS, Saquel C, Catalán RJ, et al. Axonal Degeneration Is Mediated by Necroptosis Activation. J Neurosci. 2019;39(20):3832–3844.
  • Humphries F, Yang S, Wang B, et al. RIP kinases: key decision makers in cell death and innate immunity. Cell Death Differ. 2015;22(2):225–236.
  • Qian J, Herrera JJ, Narayana PA. Neuronal and axonal degeneration in experimental spinal cord injury: in vivo proton magnetic resonance spectroscopy and histology. J Neurotrauma. 2009;27(3):599–610.
  • Kanno H, Ozawa H, Tateda S, et al. Upregulation of the receptor-interacting protein 3 expression and involvement in neural tissue damage after spinal cord injury in mice. BMC Neurosci. 2015;16(1):62.
  • Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Myelin damage and repair in pathologic CNS: challenges and prospects. Front Mol Neurosci. 2015;8.
  • Plemel JR, et al. Remyelination after spinal cord injury: is it a target for repair? Prog Neurobiol. 2014;117:54–72.
  • Su L-J, Zhang J-H, Gomez H, et al. Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid Med Cell Longev. 2019;2019:1–13.
  • Karimi-Abdolrezaee S, Eftekharpour E, Fehlings MG. Temporal and spatial patterns of Kv1.1 and Kv1.2 protein and gene expression in spinal cord white matter after acute and chronic spinal cord injury in rats: implications for axonal pathophysiology after neurotrauma. Eur J Neurosci. 2004;19(3):577–589.
  • Hunanyan AS, Alessi V, Patel S, et al. Alterations of action potentials and the localization of Nav1.6 sodium channels in spared axons after hemisection injury of the spinal cord in adult rats. J Neurophysiol. 2011;105(3):1033–1044.
  • Papastefanaki F, Matsas R. From demyelination to remyelination: the road toward therapies for spinal cord injury. Glia. 2015;63:1101–1125.
  • Oberheim NA, Goldman SA, Nedergaard M. Heterogeneity of astrocytic form and function. Methods Mol Biol Clifton NJ. 2012;814:23–45.
  • Lukovic D, Stojkovic M, Moreno-Manzano V, et al. Concise review: reactive astrocytes and stem cells in spinal cord injury: good guys or bad guys?: stem cell and astrogliosis. Stem Cells. 2015;33(4):1036–1041.
  • Muñoz-Galdeano T, Reigada D, del Águida Á, et al. Cell specific changes of autophagy in a mouse model of contusive spinal cord injury. Front Cell Neurosci. 2018;12. DOI:https://doi.org/10.3389/fncel.2018.00164.
  • Fan H, Zhang K, Shan L, et al. Reactive astrocytes undergo M1 microglia/macrohpages-induced necroptosis in spinal cord injury. Mol Neurodegener. 2016;11(1):14.
  • Lepore AC, O’Donnell J, Bonner JF, et al. Spatial and temporal changes in promoter activity of the astrocyte glutamate transporter GLT1 following traumatic spinal cord injury. J Neurosci Res. 2011;89(7):1001–1017.
  • Nicaise C, Mitrecic D, Falnikar A, et al. Transplantation of stem cell-derived astrocytes for the treatment of amyotrophic lateral sclerosis and spinal cord injury. World J Stem Cells. 2015;7(2):380–398.
  • Hara M, Kobayakawa K, Ohkawa Y, et al. Interaction of reactive astrocytes with type I collagen induces astrocytic scar formation through the integrin–N-cadherin pathway after spinal cord injury. Nat Med. 2017;23(7):818–828.
  • Li K, Javed E, Scura D, et al. Human iPS cell-derived astrocyte transplants preserve respiratory function after spinal cord injury. Exp Neurol. 2015;271:479–492.
  • Bellver-Landete V, Bretheau F, Mailhot B, et al. Microglia are an essential component of the neuroprotective scar that forms after spinal cord injury. Nat Commun. 2019;10(1):518.
  • El Tecle NE, Dahdaleh NS, Hitchon PW. Timing of surgery in spinal cord injury. Spine (Phila Pa 1976). 2016;41(16):E995–E1004.
  • Lee D-Y, Park Y-J, Song S-Y, et al. The importance of early surgical decompression for acute traumatic spinal cord injury. Clin Orthop Surg. 2018;10(4):448–454.
  • Xie J, Zhang X, Li Q, et al. Inhibition of inflammatory cytokines after early decompression may mediate recovery of neurological function in rats with spinal cord injury. Neural Regen Res. 2015;10(2):219–224.
  • Ng I, Yap E, Lim J. Changes in cerebral hemodynamics and cerebral oxygenation during surgical evacuation for hypertensive intracerebral putaminal hemorrhage. Acta Neurochir Suppl. 2005;95(5):97–101.
  • Sámano C, Nistri A. Mechanism of neuroprotection against experimental spinal cord injury by riluzole or methylprednisolone. Neurochem Res. 2019;44:200–213.
  • Yousefifard M, Rahimi-Movaghar V, Nasirinezhad F, et al. Neural stem/progenitor cell transplantation for spinal cord injury treatment; A systematic review and meta-analysis. Neuroscience. 2016;322:377–397.
  • Sandrow-Feinberg HR, Izzi J, Shumsky JS, et al. Forced exercise as a rehabilitation strategy after unilateral cervical spinal cord contusion injury. J Neurotrauma. 2009;26(5):721–731.
  • Sandrow-Feinberg HR, Houlé JD. Exercise after spinal cord injury as an agent for neuroprotection, regeneration and rehabilitation. Brain Res. 2015;1619:12–21.
  • Bobinski F, Martins DF, Bratti T, et al. Neuroprotective and neuroregenerative effects of low-intensity aerobic exercise on sciatic nerve crush injury in mice. Neuroscience. 2011;194:337–348.
  • Kumamaru H, Lu P, Rosenzweig ES, et al. Regenerating corticospinal axons innervate phenotypically appropriate neurons within neural stem cell grafts. Cell Rep. 2019;26(9):2329–2339.e4.
  • Dulin JN, Adler AF, Kumamaru H, et al. Injured adult motor and sensory axons regenerate into appropriate organotypic domains of neural progenitor grafts. Nat Commun. 2018;9(1). DOI:https://doi.org/10.1038/s41467-017-02613-x.
  • Rosenzweig ES, Brock JH, Lu P, et al. Restorative effects of human neural stem cell grafts on the primate spinal cord. Nat Med. 2018;24(4):484–490.
  • Kadoya K, Lu P, Nguyen K, et al. Spinal cord reconstitution with homologous neural grafts enables robust corticospinal regeneration. Nat Med. 2016;22:479–487.
  • Wu S, FitzGerald KT, Giordano J. On the viability and potential value of stem cells for repair and treatment of central neurotrauma: overview and speculations. Front Neurol. 2018;9:602.
  • Alonso GR. Neuronal progenitor-like cells expressing polysialylated neural cell adhesion molecule are present on the ventricular surface of the adult rat brain and spinal cord. J Comp Neurol. 1999;414(2):18.
  • Mothe AJ, Zahir T, Santaguida C, et al. Neural stem/progenitor cells from the adult human spinal cord are multipotent and self-renewing and differentiate after transplantation. PLoS ONE. 2011;6(11):e27079.
  • Zhang S-C, Wernig M, Duncan ID, et al. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol. 2001;19(12):1129–1133.
  • Chambers SM, Fasano CA, Papapetrou EP, et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27(3):275–280.
  • Han DW, Tapia N, Hermann A, et al. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell. 2012;10(4):465–472.
  • Wang L, Huang W,et al. Generation of integration-free neural progenitor cells from cells in human urine. Nat Methods. 2013;10:84–89.
  • Cassady JP, D’Alessio A, Sarkar S, et al. Direct lineage conversion of adult mouse liver cells and B lymphocytes to neural stem cells. Stem Cell Rep. 2014;3(6):948–956.
  • Lu P, Woodruff G, Wang Y, et al. Long-distance axonal growth from human induced pluripotent stem cells after spinal cord injury. Neuron. 2014;83(4):789–796.
  • Lu P, Wang Y, Graham L, et al. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell. 2012;150(6):1264–1273.
  • Bonner JF, Steward O. Repair of spinal cord injury with neuronal relays: from fetal grafts to neural stem cells. Brain Res. 2015;1619:115–123.
  • Kumamaru H, Saiwai H, Kubota K, et al. Therapeutic activities of engrafted neural stem/precursor cells are not dormant in the chronically injured spinal cord. Stem Cells. 2013;31(8):1535–1547.
  • Tscherter A, Heidemann M, Kleinlogel S, et al. Embryonic cell grafts in a culture model of spinal cord lesion: neuronal relay formation is essential for functional regeneration. Front Cell Neurosci. 2016;10. DOI:https://doi.org/10.3389/fncel.2016.00220
  • Karimi-Abdolrezaee S, Eftekharpour E, Wang J, et al. Delayed transplantation of adult neural precursor cells promotes remyelination and functional neurological recovery after spinal cord injury. J Neurosci Off J Soc Neurosci. 2006;26:3377–3389.
  • Dyck SM, Alizadeh A, Santhosh KT, et al. Chondroitin sulfate proteoglycans negatively modulate spinal cord neural precursor cells by signaling through LAR and RPTPσ and modulation of the Rho/ROCK pathway: CSPGs mechanisms in regulating spinal cord npcs. Stem Cells. 2015;33(8):2550–2563.
  • Setoguchi T, Nakashima K, Takizawa T, et al. Treatment of spinal cord injury by transplantation of fetal neural precursor cells engineered to express BMP inhibitor. Exp Neurol. 2004;189(1):33–44.
  • Wang B, Xiao Z, Chen B, et al. Nogo-66 promotes the differentiation of neural progenitors into astroglial lineage cells through mTOR-STAT3 pathway. Plos One. 2008;3(3):e1856.
  • Barnabé-Heider F, Göritz C, Sabelström H, et al. Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell. 2010;7(4):470–482.
  • Namiki J, Tator CH. Cell proliferation and nestin expression in the ependyma of the adult rat spinal cord after injury. J Neuropathol Exp Neurol. 1999;58(5):489–498.
  • Faulkner JR, Herrmann JE, Woo MJ, et al. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci Off J Soc Neurosci. 2004;24(9):2143–2155.
  • Anderson MA, Burda JE, Ren Y, et al. Astrocyte scar formation aids central nervous system axon regeneration. Nat. 2016;532(7598):195–200.
  • Davies SJA, Shih C-H, Noble M, et al. Transplantation of specific human astrocytes promotes functional recovery after spinal cord injury. PLoS ONE. 2011;6(3):e17328.
  • Hofstetter CP, Holmström NAV, Lilja JA, et al. Allodynia limits the usefulness of intraspinal neural stem cell grafts; directed differentiation improves outcome. Nat Neurosci. 2005;8:346–353.
  • Jin Y, Neuhuber B, Singh A, et al. Transplantation of human glial restricted progenitors and derived astrocytes into a contusion model of spinal cord injury. J Neurotrauma. 2011;28(4):579–594.
  • Crawford TQ, Roelink H. The notch response inhibitor DAPT enhances neuronal differentiation in embryonic stem cell-derived embryoid bodies independently of sonic hedgehog signaling. Dev Dyn. 2007;236(3):886–892.
  • Wang J, Ye Z, Zheng S, et al. Lingo-1 shRNA and Notch signaling inhibitor DAPT promote differentiation of neural stem/progenitor cells into neurons. Brain Res. 2016;1634:34–44.
  • Abematsu M, Tsujimura K, Yamano M, et al. Neurons derived from transplanted neural stem cells restore disrupted neuronal circuitry in a mouse model of spinal cord injury. J Clin Invest. 2010;120:3255–3266.
  • Wang L, Liu Y, Li S, et al. Wnt signaling pathway participates in valproic acid-induced neuronal differentiation of neural stem cells. Int J Clin Exp Pathol. 2015;8:578–585.
  • Chu T, Zhou H, Wang T, et al. In vitro characteristics of valproic acid and all-trans-retinoic acid and their combined use in promoting neuronal differentiation while suppressing astrocytic differentiation in neural stem cells. Brain Res. 2015;1596:31–47.
  • Mayer-Proschel M, Kalyani AJ, Mujtaba T, et al. Isolation of lineage-restricted neuronal precursors from multipotent neuroepithelial stem cells. Neuron. 1997;19(4):773–785.
  • Han SSW, Kang DY, Mujtaba T, et al. Grafted lineage-restricted precursors differentiate exclusively into neurons in the adult spinal cord. Exp Neurol. 2002;177(2):360–375.
  • Navarro Quiroz E, Navarro Quiroz R, Ahmad M, et al. Cell signaling in neuronal stem cells. Cells. 2018;7(7):75.
  • Blits B, Kitay BM, Farahvar A, et al. Lentiviral vector-mediated transduction of neural progenitor cells before implantation into injured spinal cord and brain to detect their migration, deliver neurotrophic factors and repair tissue. Restor Neurol Neurosci. 2005;23(5–6):313–324.
  • Butenschön J, Zimmermann T, Schmarowski N, et al. PSA-NCAM positive neural progenitors stably expressing BDNF promote functional recovery in a mouse model of spinal cord injury. Stem Cell Res Ther. 2016;7:11.
  • He B-L, Ba Y-C, Wang X-Y, et al. BDNF expression with functional improvement in transected spinal cord treated with neural stem cells in adult rats. Neuropeptides. 2013;47(1):1–7.
  • Bradbury EJ, King VR, Simmons LJ, et al. NT-3, but not BDNF, prevents atrophy and death of axotomized spinal cord projection neurons. Eur J Neurosci. 1998;10(10):3058–3068.
  • Hwang DH, Kim HM, Kang YM, et al. Combination of multifaceted strategies to maximize the therapeutic benefits of neural stem cell transplantation for spinal cord repair. Cell Transplant. 2011;20(9):1361–1379.
  • Hwang K, Jung K, Kim I, et al. Glial cell line-derived neurotrophic factor-overexpressing human neural stem/progenitor cells enhance therapeutic efficiency in rat with traumatic spinal cord injury. Exp Neurobiol. 2019;28:679–696.
  • Tiraihi T, Taheri T, Abdanipour A. Intraspinal transplantation of motoneuron-like cell combined with delivery of polymer-based glial cell line-derived neurotrophic factor for repair of spinal cord contusion injury. Neural Regen Res. 2014;9:1003.
  • Khazaei M, Ahuja CS, Nakashima H, et al.GDNF rescues the fate of neural progenitor grafts by attenuating notch signals in the injured spinal cord in rodents. Sci Transl Med. 2020;12(525):eaau3538.
  • Michelsen KA, Acosta-Verdugo S, Benoit-Marand M, et al. Area-specific reestablishment of damaged circuits in the adult cerebral cortex by cortical neurons derived from mouse embryonic stem cells. Neuron. 2015;85(5):982–997.
  • Espuny-Camacho I, Michelsen K, Gall D, et al. Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron. 2013;77(3):440–456.
  • Falkner S, Grade S, Dimou L, et al. Transplanted embryonic neurons integrate into adult neocortical circuits. Nature. 2016;539(7628):248–253.
  • Wuttke TV, Markopoulos F, Padmanabhan H, et al. Developmentally primed cortical neurons maintain fidelity of differentiation and establish appropriate functional connectivity after transplantation. Nat. Neurosci. 2018;21(4):517–529.
  • Espuny-Camacho I, Michelsen KA, Linaro D, et al. Human pluripotent stem-cell-derived cortical neurons integrate functionally into the lesioned adult murine visual cortex in an area-specific way. Cell Rep. 2018;23(9):2732–2743.
  • Ideguchi M, Palmer TD, Recht LD, et al. Murine embryonic stem cell-derived pyramidal neurons integrate into the cerebral cortex and appropriately project axons to subcortical targets. J Neurosci Off J Soc Neurosci. 2010;30(3):894–904.
  • Sagner A, Briscoe J. Establishing neuronal diversity in the spinal cord: a time and a place. Development. 2019;146(22):dev182154.
  • Ross SE, Greenberg ME, Stiles CD. Basic helix-loop-helix factors in cortical development. Neuron. 2003;39(1):13–25.
  • Lobe CG. Expression of the helix-loop-helix factor, Hes3, during embryo development suggests a role in early midbrain-hindbrain patterning. Mech Dev. 1997;62(2):227–237.
  • Duncan MK, Bordas L, Dicicco-Bloom E, et al. Expression of the helix-loop-helix genes Id-1 and NSCL-1 during cerebellar development. Dev Dyn Off Publ Am Assoc Anat. 1997;208:107–114.
  • Filippi A, Tiso N, Deflorian G, et al. The basic helix-loop-helix olig3 establishes the neural plate boundary of the trunk and is necessary for development of the dorsal spinal cord. Proc Natl Acad Sci USA. 2005;102:4377–4382.
  • CurtA, Hsieh J, Schubert M, et al. Safety and preliminary efficacy of allogeneic neural stem cell transplantation in chronic spinal cord injury: a translational phase I/IIa trial. https://papers.ssrn.com/abstract=3335037 2019.
  • Tsuji O, Sugai K, Yamaguchi R, et al. Concise review: laying the groundwork for a first-in-human study of an induced pluripotent stem cell-based intervention for spinal cord injury. Stem Cells. 2019;37(1):6–13.
  • Riemann L, Younsi A, Scherer M, et al. Transplantation of neural precursor cells attenuates chronic immune environment in cervical spinal cord injury. Front Neurol. 2018;9. DOI:https://doi.org/10.3389/fneur.2018.00428.
  • Amemori T, Romanyuk N, Jendelova P, et al. Human conditionally immortalized neural stem cells improve locomotor function after spinal cord injury in the rat. Stem Cell Res Ther. 2013;4:68.
  • Cocks G, Romanyuk N, Amemori T, et al. Conditionally immortalized stem cell lines from human spinal cord retain regional identity and generate functional V2a interneurons and motorneurons. Stem Cell Res Ther. 2013;4(3):69.
  • Ceto S, Sekiguchi KJ, Takashima Y, et al. Neural stem cell grafts form extensive synaptic networks that integrate with host circuits after spinal cord injury. Cell Stem Cell. 2020;27(3):430–440.e5.
  • Friese A, Kaltschmidt JA, Ladle DR, et al. Gamma and alpha motor neurons distinguished by expression of transcription factor Err3. Proc Natl Acad Sci. 2009;106:13588–13593.
  • Lalancette-Hebert M, Sharma A, Lyashchenko AK, et al. Gamma motor neurons survive and exacerbate alpha motor neuron degeneration in ALS. Proc Natl Acad Sci. 2016;113(51):E8316–E8325.
  • Ichiyama RM, Broman J, Roy RR, et al. Locomotor training maintains normal inhibitory influence on both alpha- and gamma-motoneurons after neonatal spinal cord transection. J Neurosci. 2011;31:26–33.
  • Grycz K, Głowacka A, Ji B, et al. Early pre- and postsynaptic decrease in glutamatergic and cholinergic signaling after spinalization is not modified when stimulating proprioceptive input to the ankle extensor α-motoneurons: anatomical and neurochemical study. PLoS ONE. 2019;14(9):e0222849.
  • Davis-Dusenbery BN, Williams LA, Klim JR, et al. How to make spinal motor neurons. Development. 2014;141(3):491–501.
  • Sabharwal P, Lee C, Park S, et al. GDE2 regulates subtype-specific motor neuron generation through inhibition of notch signaling. Neuron. 2011;71(6):1058–1070.
  • Kim JH, Shim SR, Doo SW, et al. Bladder recovery by stem cell based cell therapy in the bladder dysfunction induced by spinal cord injury: systematic review and meta-analysis. PLoS ONE. 2015;10(3):e0113491-e0113491.
  • Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci. 2020;21(7):366–383.
  • Erceg S, Ronaghi M, Oria M, et al. Transplanted oligodendrocytes and motoneuron progenitors generated from human embryonic stem cells promote locomotor recovery after spinal cord transection. Stem Cells Dayt Ohio. 2010;28(9):1541–1549.
  • Rossi SL, Nistor G, Wyatt T, et al.Histological and functional benefit following transplantation of motor neuron progenitors to the injured rat spinal cord. PloS One. 2010;5(7):e11852.
  • Trawczynski M, Liu G, David BT, et al. Restoring motor neurons in spinal cord injury with induced pluripotent stem cells. Front Cell Neurosci. 2019;13:369.
  • Goldshmit Y, Tang JKKY, Siegel AL, et al. Different Fgfs have distinct roles in regulating neurogenesis after spinal cord injury in zebrafish. Neural Dev. 2018;13(1):24.
  • Sathyamurthy A, Johnson KR, Matson KJE, et al. Massively parallel single nucleus transcriptional profiling defines spinal cord neurons and their activity during behavior. Cell Rep. 2018;22(8):2216–2225.
  • Rosenberg AB, Roco CM, Muscat RA, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science. 2018;360(6385):176–182.
  • Gupta S, Sivalingam D, Hain S, et al. Deriving dorsal spinal sensory interneurons from human pluripotent stem cells. Stem Cell Rep. 2018;10(2):390–405.
  • Zholudeva LV, Iyer N, Qiang L, et al. Transplantation of neural progenitors and V2a interneurons after spinal cord injury. J Neurotrauma. 2018;35(24):2883–2903.
  • Brown CR, Butts JC, McCreedy DA, et al. Generation of V2a interneurons from mouse embryonic stem cells. Stem Cells Dev. 2014;23(15):1765–1776.
  • Iyer NR, Huettner JE, Butts JC, et al. Generation of highly enriched V2a interneurons from mouse embryonic stem cells. Exp Neurol. 2016;277:305–316.
  • Butts JC, McCreedy DA, Martinez-Vargas JA, et al. Differentiation of V2a interneurons from human pluripotent stem cells. Proc Natl Acad Sci U. S. A. 2017;114(19):4969–4974.
  • Xu H, Sakiyama-Elbert SE. Directed differentiation of V3 interneurons from mouse embryonic stem cells. Stem Cells Dev. 2015;24(22):2723–2732.
  • Xu H, Iyer N, Huettner JE, et al. A puromycin selectable cell line for the enrichment of mouse embryonic stem cell-derived V3 interneurons. Stem Cell Res Ther. 2015;6(1). DOI:https://doi.org/10.1186/s13287-015-0213-z
  • Zhang Y, Narayan S, Geiman E, et al. V3 spinal neurons establish a robust and balanced locomotor rhythm during walking. Neuron. 2008;60(1):84–96.
  • Lin S, Li Y, Lucas-Osma AM, et al. Locomotor-related V3 interneurons initiate and coordinate muscles spasms after spinal cord injury. J Neurophysiol. 2019;121(4):1352–1367.
  • Dugan EA, Jergova S, Sagen J. Mutually beneficial effects of intensive exercise and GABAergic neural progenitor cell transplants in reducing neuropathic pain and spinal pathology in rats with spinal cord injury. Exp Neurol. 2020;327:113208.
  • Lee JW, Jergova S, Furmanski O, et al. Predifferentiated GABAergic neural precursor transplants for alleviation of dysesthetic central pain following excitotoxic spinal cord injury. Front Physiol. 2012;3. DOI:https://doi.org/10.3389/fphys.2012.00167
  • Jergova S, Hentall ID, Gajavelli S, et al. Intraspinal transplantation of GABAergic neural progenitors attenuates neuropathic pain in rats: a pharmacologic and neurophysiological evaluation. Exp Neurol. 2012;234(1):39–49.
  • Manion J, Khuong T, Harney D, et al. Human induced pluripotent stem cell-derived GABAergic interneuron transplants attenuate neuropathic pain. Pain. 2020;161(2):379–387.
  • Fandel TM, Trivedi A, Nicholas C, et al. Transplanted human stem cell-derived interneuron precursors mitigate mouse bladder dysfunction and central neuropathic pain after spinal cord injury. Cell Stem Cell. 2016;19(4):544–557.
  • Bráz JM, Wang X, Guan Z, et al. Transplant-mediated enhancement of spinal cord GABAergic inhibition reverses paclitaxel-induced mechanical and heat hypersensitivity. Pain. 2015;156(6):1084–1091.
  • Hwang I, Hahm S-C, Choi K-A, et al. Intrathecal transplantation of embryonic stem cell-derived spinal GABAergic neural precursor cells attenuates neuropathic pain in a spinal cord injury rat model. Cell Transplant. 2016;25(3):593–607.
  • Waxman SG. Demyelination in spinal cord injury. J Neurol Sci. 1989;91(1–2):1–14.
  • Assinck P, Duncan GJ, Plemel JR, et al. Myelinogenic plasticity of oligodendrocyte precursor cells following spinal cord contusion injury. J Neurosci. 2017;37(36):8635–8654.
  • Hesp ZC, Goldstein EA, Miranda CJ, et al. Chronic oligodendrogenesis and remyelination after spinal cord injury in mice and rats. J Neurosci. 2015;35(3):1274–1290.
  • Lacroix S, Hamilton LK, Vaugeois A, et al. Central canal ependymal cells proliferate extensively in response to traumatic spinal cord injury but not demyelinating lesions. PLoS ONE. 2014;9(1):e85916.
  • Shultz RB, Wang Z, Nong J, et al. Local delivery of thyroid hormone enhances oligodendrogenesis and myelination after spinal cord injury. J Neural Eng. 2017;14(3):036014.
  • Jiang S, Ballerini P, Buccella S, et al. Remyelination after chronic spinal cord injury is associated with proliferation of endogenous adult progenitor cells after systemic administration of guanosine. Purinergic Signal. 2008;4(1):61–71.
  • Whittaker MT, Zai LJ, Lee HJ, et al. GGF2 (Nrg1-β3) treatment enhances NG2+ cell response and improves functional recovery after spinal cord injury. Glia. 2012;60(2):281–294.
  • Smith DR, Margul DJ, Dumont CM, et al. Combinatorial lentiviral gene delivery of pro-oligodendrogenic factors for improving myelination of regenerating axons after spinal cord injury. Biotechnol Bioeng. 2019;116(1):155–167.
  • Powers BE, Lasiene J, Plemel JR, et al.Axonal thinning and extensive remyelination without chronic demyelination in spinal injured rats. J Neurosci. 2012;32(15):5120–5125.
  • Totoiu MO, Keirstead HS. Spinal cord injury is accompanied by chronic progressive demyelination. J Comp Neurol. 2005;486(4):373–383.
  • Kawabata S, Takano M, Numasawa-Kuroiwa Y, et al. Grafted human iPS cell-derived oligodendrocyte precursor cells contribute to robust remyelination of demyelinated axons after spinal cord injury. Stem Cell Rep. 2016;6(1):1–8.
  • Salewski RP, Mitchell RA, Li L, et al. Transplantation of induced pluripotent stem cell-derived neural stem cells mediate functional recovery following thoracic spinal cord injury through remyelination of axons: myelination following SCI with iPS-dNSC treatment. STEM CELLS Transl Med. 2015;4(7):743–754.
  • Hawryluk GWJ, Spano S, Chew D, et al. An examination of the mechanisms by which neural precursors augment recovery following spinal cord injury: a key role for remyelination. Cell Transplant. 2014;23(3):365–380.
  • Sasaki M. Molecular reconstruction of nodes of ranvier after remyelination by transplanted olfactory ensheathing cells in the demyelinated spinal cord. J Neurosci. 2006;26(6):1803–1812.
  • Eftekharpour E, Karimi-Abdolrezaee S, Wang J, et al. Myelination of congenitally dysmyelinated spinal cord axons by adult neural precursor cells results in formation of nodes of ranvier and improved axonal conduction. J Neurosci. 2007;27(13):3416–3428.
  • Katoh H, Yokota K, Fehlings MG. Regeneration of spinal cord connectivity through stem cell transplantation and biomaterial scaffolds. Front Cell Neurosci. 2019;13:248.
  • Salewski RP, Mitchell RA, Shen C, et al. Transplantation of neural stem cells clonally derived from embryonic stem cells promotes recovery after murine spinal cord injury. Stem Cells Dev. 2015;24(1):36–50.
  • Karimi-Abdolrezaee S. Delayed transplantation of adult neural precursor cells promotes remyelination and functional neurological recovery after spinal cord injury. J Neurosci. 2006;26(13):3377–3389.
  • Sankavaram SR, Hakim R, Covacu R, et al. Adult neural progenitor cells transplanted into spinal cord injury differentiate into oligodendrocytes, enhance myelination, and contribute to recovery. Stem Cell Rep. 2019;12(5):950–966.
  • Nagoshi N, Khazaei M, Ahlfors J-E, et al. Human spinal oligodendrogenic neural progenitor cells promote functional recovery after spinal cord injury by axonal remyelination and tissue sparing: oligodendrogenic NPCs for spinal cord injury. STEM CELLS Transl Med. 2018;7(11):806–818.
  • Agius E, Soukkarieh C, Danesin C, et al. Converse control of oligodendrocyte and astrocyte lineage development by sonic hedgehog in the chick spinal cord. Dev Biol. 2004;270(2):308–321.
  • Ono K, Bansal R, Payne J, et al. Early development and dispersal of oligodendrocyte precursors in the embryonic chick spinal cord. Development. 1995;121(6):12.
  • Wang S-Z. An oligodendrocyte-specific zinc-finger transcription regulator cooperates with Olig2 to promote oligodendrocyte differentiation. Development. 2006;133(17):3389–3398.
  • Biswas S, Chung SH, Jiang P, et al. Development of glial restricted human neural stem cells for oligodendrocyte differentiation in vitro and in vivo. Sci Rep. 2019;9(1):9013.
  • Goldman SA, Kuypers NJ. How to make an oligodendrocyte. Development. 2015;142(23):3983–3995.
  • Li S, Zheng J, Chai L, et al. Rapid and efficient differentiation of rodent neural stem cells into oligodendrocyte progenitor cells. Dev Neurosci. 2019;41(1–2):79–93.
  • Yamashita T, Miyamoto Y, Bando Y, et al. Differentiation of oligodendrocyte progenitor cells from dissociated monolayer and feeder-free cultured pluripotent stem cells. Plos One. 2017;12(2):e0171947.
  • Wang S, Bates J, Li X, et al. Human iPSC-derived oligodendrocyte progenitors can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell. 2013;12(2):252–264.
  • Sharp J, Frame J, Siegenthaler M, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants improve recovery after cervical spinal cord injury. Stem Cells. 2009;28:152–163.
  • Lee Y, Kim C-Y, Lee HJ, et al. Two-step generation of oligodendrocyte progenitor cells from mouse fibroblasts for spinal cord injury. Front Cell Neurosci. 2018;12:198.
  • Kim D-S, Jung SJ, Lee JS, et al. Rapid generation of OPC-like cells from human pluripotent stem cells for treating spinal cord injury. Exp Mol Med. 2017;49(7):e361–e361.
  • Priest CA, Manley NC, Denham J, et al. Preclinical safety of human embryonic stem cell-derived oligodendrocyte progenitors supporting clinical trials in spinal cord injury. Regen Med. 2015;10(8):939–958.
  • Manley NC, Priest CA, Denham J, et al. Human embryonic stem cell-derived oligodendrocyte progenitor cells: preclinical efficacy and safety in cervical spinal cord injury: hESC-derived OPCs for cervical spinal cord injury. STEM CELLS Transl Med. 2017;6(10):1917–1929.
  • Cao Q, He Q, Wang Y, et al. Transplantation of ciliary neurotrophic factor-expressing adult oligodendrocyte precursor cells promotes remyelination and functional recovery after spinal cord injury. J Neurosci. 2010;30(8):2989–3001.
  • Fan H-B, Chen L-X, Qu X-B, et al. Transplanted miR-219-overexpressing oligodendrocyte precursor cells promoted remyelination and improved functional recovery in a chronic demyelinated model. Sci Rep. 2017;7(1):41407.
  • Nazari B, Soleimani M, Ebrahimi-Barough S, et al. Overexpression of miR-219 promotes differentiation of human induced pluripotent stem cells into pre-oligodendrocyte. J Chem Neuroanat. 2018;91:8–16.
  • Chu T, Zhou H, Li F, et al. Astrocyte transplantation for spinal cord injury: current status and perspective. Brain Res Bull. 2014;107:18–30.
  • Qian D, Li L, Rong Y, et al. Blocking Notch signal pathway suppresses the activation of neurotoxic A1 astrocytes after spinal cord injury. Cell Cycle. 2019;18(21):3010–3029.
  • Shih C-H, Lacagnina M, Leuer-Bisciotti K, et al. Astroglial-derived periostin promotes axonal regeneration after spinal cord injury. J Neurosci. 2014;34(7):2438–2443.
  • Davies JE, Pröschel C, Zhang N, et al. Transplanted astrocytes derived from BMP- or CNTF-treated glial-restricted precursors have opposite effects on recovery and allodynia after spinal cord injury. J Biol. 2008;7(7):24.
  • Goulão M, Ghosh B, Urban MW, et al. Astrocyte progenitor transplantation promotes regeneration of bulbospinal respiratory axons, recovery of diaphragm function, and a reduced macrophage response following cervical spinal cord injury. Glia. 2019;67(3):452–466.
  • Li K, Javed E, Hala TJ, et al. Transplantation of glial progenitors that overexpress glutamate transporter GLT1 preserves diaphragm function following cervical SCI. Mol Ther. 2015;23(3):533–548.
  • Assinck P, Duncan GJ, Hilton BJ, et al. Cell transplantation therapy for spinal cord injury. Nat Neurosci. 2017;20(5):637–647.
  • Lane MA, Lepore AC, Fischer I. Improving the therapeutic efficacy of neural progenitor cell transplantation following spinal cord injury. Expert Rev Neurother. 2017;17:433–440.
  • Nori S, Nakamura M, Okano H. Plasticity and regeneration in the injured spinal cord after cell transplantation therapy. Prog Brain Res. 2017;231:33–56.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.