955
Views
49
CrossRef citations to date
0
Altmetric
Review

Spinal cord injuries: how could cell therapy help?

, &
Pages 529-541 | Received 08 Nov 2016, Accepted 15 Mar 2017, Published online: 27 Mar 2017

References

  • National Spinal Cord Injury Statistical Center, Facts and Figures at a Glance. Birmingham, AL: University of Alabama at Birmingham, 2016.
  • Krueger H, Noonan VK, Trenaman LM, et al. The economic burden of traumatic spinal cord injury in Canada. Chronic Dis Inj Can. 2013;33:113–122.
  • Dvorak MF, Noonan VK, Fallah N, et al. The influence of time from injury to surgery on motor recovery and length of hospital stay in acute traumatic spinal cord injury: an observational canadian cohort study. J Neurotrauma. 2015;32:645–654.
  • Wilson JR, Forgione N, Fehlings MG. Emerging therapies for acute traumatic spinal cord injury. CMAJ Can Med Assoc J J Assoc Medicale Can. 2013;185:485–492.
  • Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg. 1991;75:15–26.
  • Fehlings MG, Hawryluk GWJ. Scarring after spinal cord injury. J Neurosurg Spine. 2010;13:165–167; discussion 167–168.
  • Kanno H, Pearse DD, Ozawa H, et al. Schwann cell transplantation for spinal cord injury repair: its significant therapeutic potential and prospectus. Rev Neurosci. 2015;26:121–128.
  • Li L, Adnan H, Xu B, et al. Effects of transplantation of olfactory ensheathing cells in chronic spinal cord injury: a systematic review and meta-analysis. Eur Spine J Off Publ Eur Spine Soc Eur Spinal Deform Soc Eur Sect Cerv Spine Res Soc. 2015;24:919–930.
  • Oliveri RS, Bello S, Biering-Sørensen F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: systematic review with meta-analyses of rat models. Neurobiol Dis. 2014;62:338–353.
  • Khazaei M, Siddiqui A, Fehlings M. The potential for iPS-derived stem cells as a therapeutic strategy for spinal cord injury: opportunities and challenges. J Clin Med. 2015;4:37–65.
  • Stenudd M, Sabelström H, Frisén J. Role of endogenous neural stem cells in spinal cord injury and repair. JAMA Neurol. 2015;72:235.
  • Brockes JP, Fields KL, Raff MC. Studies on cultured rat Schwann cells. I. Establishment of purified populations from cultures of peripheral nerve. Brain Res. 1979;165:105–118.
  • Bunge MB, Wood PM. Realizing the maximum potential of Schwann cells to promote recovery from spinal cord injury. Handb Clin Neurol. 2012;109:523–540.
  • Felts PA, Smith KJ. Conduction properties of central nerve fibers remyelinated by Schwann cells. Brain Res. 1992;574:178–192.
  • Honmou O, Felts PA, Waxman SG, et al. Restoration of normal conduction properties in demyelinated spinal cord axons in the adult rat by transplantation of exogenous Schwann cells. J Neurosci Off J Soc Neurosci. 1996;16:3199–3208.
  • Bunge MB. Efficacy of Schwann cell transplantation for spinal cord repair is improved with combinatorial strategies. J Physiol. 2016;594:3533–3538.
  • Saberi H, Moshayedi P, Aghayan H-R, et al. Treatment of chronic thoracic spinal cord injury patients with autologous Schwann cell transplantation: an interim report on safety considerations and possible outcomes. Neurosci Lett. 2008;443:46–50.
  • Saberi H, Firouzi M, Habibi Z, et al. Safety of intramedullary Schwann cell transplantation for postrehabilitation spinal cord injuries: 2-year follow-up of 33 cases. J Neurosurg Spine. 2011;15:515–525.
  • Barraud P, Seferiadis AA, Tyson LD, et al. Neural crest origin of olfactory ensheathing glia. Proc Natl Acad Sci U S A. 2010;107:21040–21045.
  • Doucette JR, Kiernan JA, Flumerfelt BA. The re-innervation of olfactory glomeruli following transection of primary olfactory axons in the central or peripheral nervous system. J Anat. 1983;137(Pt 1):1–19.
  • Roet KCD, Verhaagen J. Understanding the neural repair-promoting properties of olfactory ensheathing cells. Exp Neurol. 2014;261:594–609.
  • Watzlawick R, Rind J, Sena ES, et al. Olfactory ensheathing cell transplantation in experimental spinal cord injury: effect size and reporting bias of 62 experimental treatments: a systematic review and meta-analysis. Plos Biol. 2016;14:e1002468.
  • Feron F. Autologous olfactory ensheathing cell transplantation in human spinal cord injury. Brain. 2005;128:2951–2960.
  • Mackay-Sim A, Féron F, Cochrane J, et al. Autologous olfactory ensheathing cell transplantation in human paraplegia: a 3-year clinical trial. Brain J Neurol. 2008;131:2376–2386.
  • Tabakow P, Jarmundowicz W, Czapiga B, et al. Transplantation of autologous olfactory ensheathing cells in complete human spinal cord injury. Cell Transplant. 2013;22:1591–1612.
  • Rao Y, Zhu W, Guo Y, et al. Long-term outcome of olfactory ensheathing cell transplantation in six patients with chronic complete spinal cord injury. Cell Transplant. 2013;22(Suppl 1):S21–S25.
  • Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The international society for cellular therapy position statement. Cytotherapy. 2006;8:315–317.
  • Crisan M, Yap S, Casteilla L, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–313.
  • Vawda R, Fehlings MG. Mesenchymal cells in the treatment of spinal cord injury: current & future perspectives. Curr Stem Cell Res Ther. 2013;8:25–38.
  • Badner A, Vawda R, Laliberte A, et al. Early intravenous delivery of human brain stromal cells modulates systemic inflammation and leads to vasoprotection in traumatic spinal cord injury. Stem Cells Transl Med. 2016;5:991–1003.
  • Hoogduijn MJ, Roemeling-van Rhijn M, Engela AU, et al. Mesenchymal stem cells induce an inflammatory response after intravenous infusion. Stem Cells Dev. 2013;22:2825–2835.
  • Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, et al. A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma. 2011;28:1611–1682.
  • Tolar J, Le Blanc K, Keating A, et al. Concise review: hitting the right spot with mesenchymal stromal cells. Stem Cells Dayt Ohio. 2010;28:1446–1455.
  • Karamouzian S, Nematollahi-Mahani SN, Nakhaee N, et al. Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clin Neurol Neurosurg. 2012;114:935–939.
  • Saito F, Nakatani T, Iwase M, et al. Administration of cultured autologous bone marrow stromal cells into cerebrospinal fluid in spinal injury patients: a pilot study. Restor Neurol Neurosci. 2012;30:127–136.
  • Kakabadze Z, Kipshidze N, Mardaleishvili K, et al. Phase 1 trial of autologous bone marrow stem cell transplantation in patients with spinal cord injury. Stem Cells Int. 2016;2016:6768274.
  • Vaquero J, Zurita M, Rico MA, et al. An approach to personalized cell therapy in chronic complete paraplegia: the Puerta de Hierro phase I/II clinical trial. Cytotherapy. 2016;18:1025–1036.
  • Geffner LF, Santacruz P, Izurieta M, et al. Administration of autologous bone marrow stem cells into spinal cord injury patients via multiple routes is safe and improves their quality of life: comprehensive case studies. Cell Transplant. 2008;17:1277–1293.
  • Vaquero J, Zurita M, Rico MA, et al. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy. 2017;19:349–359.
  • Weiss S, Dunne C, Hewson J, et al. Multipotent CNS stem cells are present in the adult mammalian spinal cord and ventricular neuroaxis. J Neurosci. 1996;16:7599–7609.
  • Gage FH. Mammalian Neural Stem Cells. Science. 2000;287:1433–1438.
  • Emgård M, Piao J, Aineskog H, et al. Neuroprotective effects of human spinal cord-derived neural precursor cells after transplantation to the injured spinal cord. Exp Neurol. 2014;253:138–145.
  • Parr AM, Kulbatski I, Zahir T, et al. Transplanted adult spinal cord-derived neural stem/progenitor cells promote early functional recovery after rat spinal cord injury. Neuroscience. 2008;155:760–770.
  • Gritti A, Frölichsthal-Schoeller P, Galli R, et al. Epidermal and fibroblast growth factors behave as mitogenic regulators for a single multipotent stem cell-like population from the subventricular region of the adult mouse forebrain. J Neurosci Off J Soc Neurosci. 1999;19:3287–3297.
  • 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 Off J Soc Neurosci. 2007;27:3416–3428.
  • Mothe AJ, Tator CH. Transplanted neural stem/progenitor cells generate myelinating oligodendrocytes and Schwann cells in spinal cord demyelination and dysmyelination. Exp Neurol. 2008;213:176–190.
  • Karimi-Abdolrezaee S. 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.
  • Karimi-Abdolrezaee S, Eftekharpour E, Wang J, et al. Synergistic effects of transplanted adult neural stem/progenitor cells, chondroitinase, and growth factors promote functional repair and plasticity of the chronically injured spinal cord. J Neurosci Off J Soc Neurosci. 2010;30:1657–1676.
  • Emgård M, Holmberg L, Samuelsson E-B, et al. Human neural precursor cells continue to proliferate and exhibit low cell death after transplantation to the injured rat spinal cord. Brain Res. 2009;1278:15–26.
  • 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;23:365–380.
  • Yan J, Xu L, Welsh AM, et al. Extensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cord. Plos Med. 2007;4:e39.
  • Lu P, Jones LL, Snyder EY, et al. Neural stem cells constitutively secrete neurotrophic factors and promote extensive host axonal growth after spinal cord injury. Exp Neurol. 2003;181:115–129.
  • Hawryluk GWJ, Mothe A, Wang J, et al. An in vivo characterization of trophic factor production following neural precursor cell or bone marrow stromal cell transplantation for spinal cord injury. Stem Cells Dev. 2012;21:2222–2238.
  • Martino G, Pluchino S. The therapeutic potential of neural stem cells. Nat Rev Neurosci. 2006;7:395–406.
  • 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.
  • Glass JD, Boulis NM, Johe K, et al. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: results of a phase I trial in 12 patients. Stem Cells Dayt Ohio. 2012;30:1144–1151.
  • Feldman EL, Boulis NM, Hur J, et al. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: phase 1 trial outcomes. Ann Neurol. 2014;75:363–373.
  • Levine JM, Reynolds R, Fawcett JW. The oligodendrocyte precursor cell in health and disease. Trends Neurosci. 2001;24:39–47.
  • Sabelström H, Stenudd M, Frisén J. Neural stem cells in the adult spinal cord. Exp Neurol. 2014;260:44–49.
  • Watanabe M, Toyama Y, Nishiyama A. Differentiation of proliferated NG2-positive glial progenitor cells in a remyelinating lesion. J Neurosci Res. 2002;69:826–836.
  • 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 Dayt Ohio. 2010;28:152–163.
  • Zhang YW, Denham J, Thies RS. Oligodendrocyte progenitor cells derived from human embryonic stem cells express neurotrophic factors. Stem Cells Dev. 2006;15:943–952.
  • Koch P, Opitz T, Steinbeck JA, et al. A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. Proc Natl Acad Sci U S A. 2009;106:3225–3230.
  • Shin S, Mitalipova M, Noggle S, et al. Long-term proliferation of human embryonic stem cell-derived neuroepithelial cells using defined adherent culture conditions. Stem Cells Dayt Ohio. 2006;24:125–138.
  • Rowland JW, Lee JJ, Salewski RP, et al. Generation of neural stem cells from embryonic stem cells using the default mechanism: in vitro and in vivo characterization. Stem Cells Dev. 2011;20:1829–1845.
  • 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;24:36–50.
  • 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 Off J Soc Neurosci. 2005;25:4694–4705.
  • Iwai H, Shimada H, Nishimura S, et al. Allogeneic neural stem/progenitor cells derived from embryonic stem cells promote functional recovery after transplantation into injured spinal cord of nonhuman primates. Stem Cells Transl Med. 2015;4:708–719.
  • 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;25:593–607.
  • Lukovic D, Stojkovic M, Moreno-Manzano V, et al. Perspectives and future directions of human pluripotent stem cell-based therapies: lessons from Geron’s clinical trial for spinal cord injury. Stem Cells Dev. 2014;23:1–4.
  • Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872.
  • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676.
  • Park IH, Zhao R, West JA, et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature. 2008:10(451):141–146.
  • Maekawa M, Yamaguchi K, Nakamura T, et al. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature. 2011;474:225–229.
  • Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–1920.
  • Kim K, Doi A, Wen B, et al. Epigenetic memory in induced pluripotent stem cells. Nature. 2010;467:285–290.
  • Ma H, Morey R, O’Neil RC, et al. Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature. 2014;511:177–183.
  • Zhou W, Freed CR. Adenoviral gene delivery can reprogram human fibroblasts to induced pluripotent stem cells. Stem Cells. 2009;27:2667–2674.
  • Ban H, Nishishita N, Fusaki N, et al. Efficient generation of transgene-free human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors. Proc Natl Acad Sci U S A. 2011;108:14234–14239.
  • Woltjen K, Michael IP, Mohseni P, et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 2009;458:766–770.
  • Yu J, Hu K, Smuga-Otto K, et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324:797–801.
  • Kim D, Kim C-H, Moon J-I, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell. 2009;4:472–476.
  • Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010;7:618–630.
  • Subramanyam D, Lamouille S, Judson RL, et al. Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat Biotechnol. 2011;29:443–448.
  • Huangfu D, Maehr R, Guo W, et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 2008;26:795–797.
  • Clancy JL, Patel HR, Hussein SMI, et al. Small RNA changes en route to distinct cellular states of induced pluripotency. Nat Commun. 2014;5:5522.
  • Salewski RP, Buttigieg J, Mitchell RA, et al. The generation of definitive neural stem cells from PiggyBac transposon-induced pluripotent stem cells can be enhanced by induction of the NOTCH signaling pathway. Stem Cells Dev. 2013;22:383–396.
  • Okubo T, Iwanami A, Kohyama J, et al. Pretreatment with a γ-secretase inhibitor prevents tumor-like overgrowth in human iPSC-derived transplants for spinal cord injury. Stem Cell Rep. 2016;7:649–663.
  • 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. Stem Cells Transl Med. 2015;4:743–754.
  • 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;6:1–8.
  • Pomeshchik Y, Puttonen KA, Kidin I, et al. Transplanted human induced pluripotent stem cell-derived neural progenitor cells do not promote functional recovery of pharmacologically immunosuppressed mice with contusion spinal cord injury. Cell Transplant. 2015;24:1799–1812.
  • Nutt SE, Chang E-A, Suhr ST, et al. Caudalized human iPSC-derived neural progenitor cells produce neurons and glia but fail to restore function in an early chronic spinal cord injury model. Exp Neurol. 2013;248:491–503.
  • López-Serrano C, Torres-Espín A, Hernández J, et al. Effects of the spinal cord injury environment on the differentiation capacity of human neural stem cells derived from induced pluripotent stem cells. Cell Transplant. 2016;25:1833–1852.
  • Campbell A, Brieva T, Raviv L, et al. Concise review: process development considerations for cell therapy. Stem Cells Transl Med. 2015;4:1155–1163.
  • Nori S, Okada Y, Nishimura S, et al. Long-term safety issues of iPSC-based cell therapy in a spinal cord injury model: oncogenic transformation with epithelial-mesenchymal transition. Stem Cell Rep. 2015;4:360–373.
  • Sotiropoulou PA, Perez SA, Salagianni M, et al. Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells Dayt Ohio. 2006;24:462–471.
  • McEwen KR, Leitch HG, Amouroux R, et al. The impact of culture on epigenetic properties of pluripotent stem cells and pre-implantation embryos. Biochem Soc Trans. 2013;41:711–719.
  • Bentivegna A, Miloso M, Riva G, et al. DNA methylation changes during in vitro propagation of human mesenchymal stem cells: implications for their genomic stability? Stem Cells Int. 2013;2013:1–9.
  • Chen X, Chen A, Woo TL, et al. Investigations into the metabolism of two-dimensional colony and suspended microcarrier cultures of human embryonic stem cells in serum-free media. Stem Cells Dev. 2010;19:1781–1792.
  • Sun N, Longaker MT, Wu JC. Human iPS cell-based therapy: considerations before clinical applications. Cell Cycle. 2010;9:880–885.
  • Unger C, Gao S, Cohen M, et al. Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Hum Reprod Oxf Engl. 2009;24:2567–2581.
  • Moll G, Alm JJ, Davies LC, et al. Do cryopreserved mesenchymal stromal cells display impaired immunomodulatory and therapeutic properties? Stem Cells. 2014;32:2430–2442.
  • François M, Copland IB, Yuan S, et al. Cryopreserved mesenchymal stromal cells display impaired immunosuppressive properties as a result of heat-shock response and impaired interferon-γ licensing. Cytotherapy. 2012;14:147–152.
  • Sohn HS, Heo JS, Kim H-S, et al. Duration of in vitro storage affects the key stem cell features of human bone marrow-derived mesenchymal stromal cells for clinical transplantation. Cytotherapy. 2013;15:460–466.
  • Chen Y, Yu B, Xue G, et al. Effects of storage solutions on the viability of human umbilical cord mesenchymal stem cells for transplantation. Cell Transplant. 2013;22:1075–1086.
  • Bradley JA, Bolton EM, Pedersen RA. Stem cell medicine encounters the immune system. Nat Rev Immunol. 2002;2:859–871.
  • Ensor CR, Trofe-Clark J, Gabardi S, et al. Generic maintenance immunosuppression in solid organ transplant recipients. Pharmacotherapy. 2011;31:1111–1129.
  • Hunt J, Cheng A, Hoyles A, et al. Cyclosporin A has direct effects on adult neural precursor cells. J Neurosci Off J Soc Neurosci. 2010;30:2888–2896.
  • Song LH, Pan W, Yu YH, et al. Resveratrol prevents CsA inhibition of proliferation and osteoblastic differentiation of mouse bone marrow-derived mesenchymal stem cells through an ER/NO/cGMP pathway. Toxicol Vitro Int J Publ Assoc BIBRA. 2006;20:915–922.
  • Sontag CJ, Nguyen HX, Kamei N, et al. Immunosuppressants affect human neural stem cells in vitro but not in an in vivo model of spinal cord injury. Stem Cells Transl Med. 2013;2:731–744.
  • Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32:252–260.
  • Chen Y-T, Yang -C-C, Zhen -Y-Y, et al. Cyclosporine-assisted adipose-derived mesenchymal stem cell therapy to mitigate acute kidney ischemia-reperfusion injury. Stem Cell Res Ther. 2013;4:62.
  • 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.
  • Ide C, Kanekiyo K. Points regarding cell transplantation for the treatment of spinal cord injury. Neural Regen Res. 2016;11:1046–1049.
  • Wu S, Cui G, Shao H, et al. The cotransplantation of olfactory ensheathing cells with bone marrow mesenchymal stem cells exerts antiapoptotic effects in adult rats after spinal cord injury. Stem Cells Int. 2015;2015:1–13.
  • Ge L, Liu K, Liu Z, et al. Co-transplantation of autologous OM-MSCs and OM-OECs: a novel approach for spinal cord injury. Rev Neurosci. 2016;27:259–270.
  • Oh JS, Kim KN, An SS, et al. Cotransplantation of mouse neural stem cells (mNSCs) with adipose tissue-derived mesenchymal stem cells improves mNSC survival in a rat spinal cord injury model. Cell Transplant. 2011;20:837–849.
  • Yazdani SO, Hafizi M, Zali A-R, et al. Safety and possible outcome assessment of autologous Schwann cell and bone marrow mesenchymal stromal cell co-transplantation for treatment of patients with chronic spinal cord injury. Cytotherapy. 2013;15:782–791.
  • Oraee-Yazdani S, Hafizi M, Atashi A, et al. Co-transplantation of autologous bone marrow mesenchymal stem cells and Schwann cells through cerebral spinal fluid for the treatment of patients with chronic spinal cord injury: safety and possible outcome. Spinal Cord. 2016;54:102–109.
  • Zeng Y-S, Ding Y, Wu L-Z, et al. Co-transplantation of Schwann cells promotes the survival and differentiation of neural stem cells transplanted into the injured spinal cord. Dev Neurosci. 2005;27:20–26.
  • Niapour A, Karamali F, Nemati S, et al. Cotransplantation of human embryonic stem cell-derived neural progenitors and Schwann cells in a rat spinal cord contusion injury model elicits a distinct neurogenesis and functional recovery. Cell Transplant. 2012;21:827–843.
  • Zhang J, Chen H, Duan Z, et al. The effects of co-transplantation of olfactory ensheathing cells and Schwann cells on local inflammation environment in the contused spinal cord of rats. Mol Neurobiol. 2017;54(2):943–953.
  • Pearse DD, Sanchez AR, Pereira FC, et al. Transplantation of Schwann cells and/or olfactory ensheathing glia into the contused spinal cord: survival, migration, axon association, and functional recovery. Glia. 2007;55:976–1000.
  • Hu J-G, Wang X-F, Deng L-X, et al. Cotransplantation of glial restricted precursor cells and Schwann cells promotes functional recovery after spinal cord injury. Cell Transplant. 2013;22:2219–2236.
  • Kanno H, Pressman Y, Moody A, et al. Combination of engineered Schwann cell grafts to secrete neurotrophin and chondroitinase promotes axonal regeneration and locomotion after spinal cord injury. J Neurosci Off J Soc Neurosci. 2014;34:1838–1855.
  • Lee SH, Kim Y, Rhew D, et al. Impact of local injection of brain-derived neurotrophic factor-expressing mesenchymal stromal cells (MSCs) combined with intravenous MSC delivery in a canine model of chronic spinal cord injury. Cytotherapy. 2016 Oct 28. doi: 10.1016/j.jcyt.2016.09.014.
  • Abbaszadeh H-A, Tiraihi T, Noori-Zadeh A, et al. Human ciliary neurotrophic factor-overexpressing stable bone marrow stromal cells in the treatment of a rat model of traumatic spinal cord injury. Cytotherapy. 2015;17:912–921.
  • Dooley D, Lemmens E, Vangansewinkel T, et al. Cell-based delivery of Interleukin-13 directs alternative activation of macrophages resulting in improved functional outcome after spinal cord injury. Stem Cell Rep. 2016;7:1099–1115.
  • Quaranta P, Focosi D, Di Iesu M, et al. Human Wharton’s jelly-derived mesenchymal stromal cells engineered to secrete Epstein-Barr virus interleukin-10 show enhanced immunosuppressive properties. Cytotherapy. 2016;18:205–218.
  • Guo J-S, Zeng Y-S, Li H-B, et al. Cotransplant of neural stem cells and NT-3 gene modified Schwann cells promote the recovery of transected spinal cord injury. Spinal Cord. 2007;45:15–24.
  • Pan H-C, Cheng F-C, Lai S-Z, et al. Enhanced regeneration in spinal cord injury by concomitant treatment with granulocyte colony-stimulating factor and neuronal stem cells. J Clin Neurosci. 2008;15:656–664.
  • 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 Off J Soc Neurosci. 2010;30:2989–3001.
  • Yoon SH, Shim YS, Park YH, et al. Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage-colony stimulating factor: phase I/II clinical trial. Stem Cells. 2007;25:2066–2073.
  • Evaniew N, Belley-Côté EP, Fallah N, et al. Methylprednisolone for the treatment of patients with acute spinal cord injuries: a systematic review and meta-analysis. J Neurotrauma. 2016;33:468–481.
  • Ahuja CS, Martin AR, Fehlings M. Recent advances in managing a spinal cord injury secondary to trauma. F1000Res. 2016;5:1017.
  • Pearse DD, Pereira FC, Marcillo AE, et al. cAMP and Schwann cells promote axonal growth and functional recovery after spinal cord injury. Nat Med. 2004;10:610–616.
  • Wang D, Zhang J. Effects of hypothermia combined with neural stem cell transplantation on recovery of neurological function in rats with spinal cord injury. Mol Med Rep. 2015;11:1759–1767.
  • Chen D, Zeng W, Fu Y, et al. Bone marrow mesenchymal stem cells combined with minocycline improve spinal cord injury in a rat model. Int J Clin Exp Pathol. 2015;8:11957–11969.
  • Iwasaki M, Wilcox JT, Nishimura Y, et al. Synergistic effects of self-assembling peptide and neural stem/progenitor cells to promote tissue repair and forelimb functional recovery in cervical spinal cord injury. Biomaterials. 2014;35:2617–2629.
  • Zweckberger K, Ahuja CS, Liu Y, et al. Self-assembling peptides optimize the post-traumatic milieu and synergistically enhance the effects of neural stem cell therapy after cervical spinal cord injury. Acta Biomater. 2016;42:77–89.
  • Mothe AJ, Tam RY, Zahir T, et al. Repair of the injured spinal cord by transplantation of neural stem cells in a hyaluronan-based hydrogel. Biomaterials. 2013;34:3775–3783.
  • Führmann T, Tam RY, Ballarin B, et al. Injectable hydrogel promotes early survival of induced pluripotent stem cell-derived oligodendrocytes and attenuates longterm teratoma formation in a spinal cord injury model. Biomaterials. 2016;83:23–36.
  • He L, Zhang Y, Zeng C, et al. Manufacture of PLGA multiple-channel conduits with precise hierarchical pore architectures and in vitro/vivo evaluation for spinal cord injury. Tissue Eng Part C Methods. 2009;15:243–255.
  • Zeng X, Zeng Y, Ma Y, et al. Bone marrow mesenchymal stem cells in a three-dimensional gelatin sponge scaffold attenuate inflammation, promote angiogenesis, and reduce cavity formation in experimental spinal cord injury. Cell Transplant. 2011;20:1881–1899.
  • Liu J, Chen J, Liu B, et al. Acellular spinal cord scaffold seeded with mesenchymal stem cells promotes long-distance axon regeneration and functional recovery in spinal cord injured rats. J Neurol Sci. 2013;325:127–136.
  • Hejcl A, Sedý J, Kapcalová M, et al. HPMA-RGD hydrogels seeded with mesenchymal stem cells improve functional outcome in chronic spinal cord injury. Stem Cells Dev. 2010;19:1535–1546.
  • Gao M, Lu P, Bednark B, et al. Templated agarose scaffolds for the support of motor axon regeneration into sites of complete spinal cord transection. Biomaterials. 2013;34:1529–1536.
  • Shrestha B, Coykendall K, Li Y, et al. Repair of injured spinal cord using biomaterial scaffolds and stem cells. Stem Cell Res Ther. 2014;5:91.
  • Houlé JD, Côté M-P. Axon regeneration and exercise-dependent plasticity after spinal cord injury. Ann N Y Acad Sci. 2013;1279:154–163.
  • Foret A, Quertainmont R, Botman O, et al. Stem cells in the adult rat spinal cord: plasticity after injury and treadmill training exercise. J Neurochem. 2010;112:762–772.
  • Tashiro S, Nishimura S, Iwai H, et al. Functional recovery from neural stem/progenitor cell transplantation combined with treadmill training in mice with chronic spinal cord injury. Sci Rep. 2016;6:30898.
  • Carvalho KAT, Cunha RC, Vialle EN, et al. Functional outcome of bone marrow stem cells (CD45(+)/CD34(-)) after cell therapy in acute spinal cord injury: in exercise training and in sedentary rats. Transplant Proc. 2008;40:847–849.
  • Yoshihara H, Shumsky JS, Neuhuber B, et al. Combining motor training with transplantation of rat bone marrow stromal cells does not improve repair or recovery in rats with thoracic contusion injuries. Brain Res. 2006;1119:65–75.
  • Sun T, Ye C, Zhang Z, et al. Cotransplantation of olfactory ensheathing cells and Schwann cells combined with treadmill training promotes functional recovery in rats with contused spinal cords. Cell Transplant. 2013;22(Suppl 1):S27–S38.
  • Dominici M, Nichols K, Srivastava A, et al. Positioning a scientific community on unproven cellular therapies: the 2015 international society for cellular therapy perspective. Cytotherapy. 2015;17:1663–1666.
  • Califf RM, Zarin DA, Kramer JM, et al. Characteristics of clinical trials registered in ClinicalTrials.gov, 2007-2010. Jama. 2012;307:1838–1847.
  • Fawcett JW, Curt A, Steeves JD, et al. Guidelines for the conduct of clinical trials for spinal cord injury as developed by the ICCP panel: spontaneous recovery after spinal cord injury and statistical power needed for therapeutic clinical trials. Spinal Cord. 2007;45:190–205.
  • Tuszynski MH, Steeves JD, Fawcett JW, et al. Guidelines for the conduct of clinical trials for spinal cord injury as developed by the ICCP Panel: clinical trial inclusion/exclusion criteria and ethics. Spinal Cord. 2007;45:222–231.
  • Daley GQ, Hyun I, Apperley JF, et al. Setting global standards for stem cell research and clinical translation: the 2016 ISSCR guidelines. Stem Cell Rep. 2016;6:787–797.

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.