Generic placeholder image

Current Stem Cell Research & Therapy

Editor-in-Chief

ISSN (Print): 1574-888X
ISSN (Online): 2212-3946

Review Article

Stem Cell Transplantation: A Promising Therapy for Spinal Cord Injury

Author(s): Zhe Gong, Kaishun Xia, Ankai Xu, Chao Yu, Chenggui Wang, Jian Zhu, Xianpeng Huang, QiXin Chen*, Fangcai Li* and Chengzhen Liang*

Volume 15, Issue 4, 2020

Page: [321 - 331] Pages: 11

DOI: 10.2174/1574888X14666190823144424

Price: $65

Abstract

Spinal Cord Injury (SCI) causes irreversible functional loss of the affected population. The incidence of SCI keeps increasing, resulting in huge burden on the society. The pathogenesis of SCI involves neuron death and exotic reaction, which could impede neuron regeneration. In clinic, the limited regenerative capacity of endogenous cells after SCI is a major problem. Recent studies have demonstrated that a variety of stem cells such as induced Pluripotent Stem Cells (iPSCs), Embryonic Stem Cells (ESCs), Mesenchymal Stem Cells (MSCs) and Neural Progenitor Cells (NPCs) /Neural Stem Cells (NSCs) have therapeutic potential for SCI. However, the efficacy and safety of these stem cellbased therapy for SCI remain controversial. In this review, we introduce the pathogenesis of SCI, summarize the current status of the application of these stem cells in SCI repair, and discuss possible mechanisms responsible for functional recovery of SCI after stem cell transplantation. Finally, we highlight several areas for further exploitation of stem cells as a promising regenerative therapy of SCI.

Keywords: Chondrogenic differentiation, adipogenic differentiation, neurons, endothelial cells, schwann cells, nucleus, pulposus- like cells, cardiomyocytes, alveolar epithelial cells.

[1]
Nakhjavan-Shahraki B, Yousefifard M, Rahimi-Movaghar V, et al. Transplantation of olfactory ensheathing cells on functional recovery and neuropathic pain after spinal cord injury; systematic review and meta-analysis. Sci Rep 2018; 8(1): 325.
[http://dx.doi.org/10.1038/s41598-017-18754-4] [PMID: 29321494]
[2]
Siebert JR, Eade AM, Osterhout DJ. Biomaterial Approaches to Enhancing Neurorestoration after Spinal Cord Injury: Strategies for Overcoming Inherent Biological Obstacles. BioMed Res Int 2015; 2015, 752572
[http://dx.doi.org/10.1155/2015/752572] [PMID: 26491685]
[3]
Doulames VM, Plant GW. Induced Pluripotent Stem Cell Therapies for Cervical Spinal Cord Injury. Int J Mol Sci 2016; 17(4): 530.
[http://dx.doi.org/10.3390/ijms17040530] [PMID: 27070598]
[4]
Hewson DW, Bedforth NM, Hardman JG. Spinal cord injury arising in anaesthesia practice. Anaesthesia 2018; 73(Suppl. 1): 43-50.
[http://dx.doi.org/10.1111/anae.14139] [PMID: 29313911]
[5]
Lukovic D, Moreno Manzano V, Stojkovic M, Bhattacharya SS, Erceg S. Concise review: human pluripotent stem cells in the treatment of spinal cord injury. Stem Cells 2012; 30(9): 1787-92.
[http://dx.doi.org/10.1002/stem.1159] [PMID: 22736576]
[6]
Baptiste DC, Fehlings MG. Pharmacological approaches to repair the injured spinal cord. J Neurotrauma 2006; 23(3-4): 318-34.
[http://dx.doi.org/10.1089/neu.2006.23.318] [PMID: 16629619]
[7]
Austin JW, Fehlings MG. Molecular mechanisms of Fas-mediated cell death in oligodendrocytes. J Neurotrauma 2008; 25(5): 411-26.
[http://dx.doi.org/10.1089/neu.2007.0436] [PMID: 18435595]
[8]
Rowland JW, Hawryluk GW, Kwon B, Fehlings MG. Current status of acute spinal cord injury pathophysiology and emerging therapies: promise on the horizon. Neurosurg Focus 2008; 25(5), E2
[http://dx.doi.org/10.3171/FOC.2008.25.11.E2] [PMID: 18980476]
[9]
Liu SM, Xiao ZF, Li X, et al. Vascular endothelial growth factor activates neural stem cells through epidermal growth factor receptor signal after spinal cord injury. CNS Neurosci Ther 2018.
[PMID: 30155986]
[10]
Chen NF, Sung CS, Wen ZH, et al. Therapeutic effect of platelet-rich plasma in rat spinal cord injuries. Front Neurosci 2018; 12: 252.
[http://dx.doi.org/10.3389/fnins.2018.00252] [PMID: 29740270]
[11]
Pires LR, Pêgo AP. Bridging the lesion-engineering a permissive substrate for nerve regeneration. Regen Biomater 2015; 2(3): 203-14.
[http://dx.doi.org/10.1093/rb/rbv012] [PMID: 26816642]
[12]
Puri MC, Nagy A. Concise review: Embryonic stem cells versus induced pluripotent stem cells: the game is on. Stem Cells 2012; 30(1): 10-4.
[http://dx.doi.org/10.1002/stem.788] [PMID: 22102565]
[13]
Nagoshi N, Okano H. Applications of induced pluripotent stem cell technologies in spinal cord injury. J Neurochem 2017; 141(6): 848-60.
[http://dx.doi.org/10.1111/jnc.13986] [PMID: 28199003]
[14]
Kramer AS, Harvey AR, Plant GW, Hodgetts SI. Systematic review of induced pluripotent stem cell technology as a potential clinical therapy for spinal cord injury. Cell Transplant 2013; 22(4): 571-617.
[http://dx.doi.org/10.3727/096368912X655208] [PMID: 22944020]
[15]
Forostyak S, Jendelova P, Sykova E. The role of mesenchymal stromal cells in spinal cord injury, regenerative medicine and possible clinical applications. Biochimie 2013; 95(12): 2257-70.
[http://dx.doi.org/10.1016/j.biochi.2013.08.004] [PMID: 23994163]
[16]
Bican O, Minagar A, Pruitt AA. The spinal cord: a review of functional neuroanatomy. Neurol Clin 2013; 31(1): 1-18.
[http://dx.doi.org/10.1016/j.ncl.2012.09.009] [PMID: 23186894]
[17]
Cao QL, Zhang YP, Howard RM, Walters WM, Tsoulfas P, Whittemore SR. Pluripotent stem cells engrafted into the normal or lesioned adult rat spinal cord are restricted to a glial lineage. Exp Neurol 2001; 167(1): 48-58.
[http://dx.doi.org/10.1006/exnr.2000.7536] [PMID: 11161592]
[18]
Ogawa Y, Sawamoto K, Miyata T, et al. Transplantation of in vitro-expanded fetal neural progenitor cells results in neurogenesis and functional recovery after spinal cord contusion injury in adult rats. J Neurosci Res 2002; 69(6): 925-33.
[http://dx.doi.org/10.1002/jnr.10341] [PMID: 12205685]
[19]
Nakamura M, Okano H. Cell transplantation therapies for spinal cord injury focusing on induced pluripotent stem cells. Cell Res 2013; 23(1): 70-80.
[http://dx.doi.org/10.1038/cr.2012.171] [PMID: 23229514]
[20]
Stenudd M, Sabelström H, Frisén J. Role of endogenous neural stem cells in spinal cord injury and repair. JAMA Neurol 2015; 72(2): 235-7.
[http://dx.doi.org/10.1001/jamaneurol.2014.2927] [PMID: 25531583]
[21]
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: 338-53.
[http://dx.doi.org/10.1016/j.nbd.2013.10.014] [PMID: 24148857]
[22]
Dalous J, Larghero J, Baud O. Transplantation of umbilical cord-derived mesenchymal stem cells as a novel strategy to protect the central nervous system: technical aspects, preclinical studies, and clinical perspectives. Pediatr Res 2012; 71(4 Pt 2): 482-90.
[http://dx.doi.org/10.1038/pr.2011.67] [PMID: 22430384]
[23]
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): 789-96.
[http://dx.doi.org/10.1016/j.neuron.2014.07.014] [PMID: 25123310]
[24]
Matsukura Y, Muneta T, Tsuji K, Koga H, Sekiya I. Mesenchymal stem cells in synovial fluid increase after meniscus injury. Clin Orthop Relat Res 2014; 472(5): 1357-64.
[http://dx.doi.org/10.1007/s11999-013-3418-4] [PMID: 24338094]
[25]
Frontini-Lopez YR, Gojanovich AD, Masone D, Bustos DM, Uhart M. Adipose-derived mesenchymal stem/stromal cells: from the lab bench to the basic concepts for clinical translation. Biocell 2018; 42(3): 67-77.
[26]
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(4): 315-7.
[http://dx.doi.org/10.1080/14653240600855905] [PMID: 16923606]
[27]
Blondheim NR, Levy YS, Ben-Zur T, et al. Human mesenchymal stem cells express neural genes, suggesting a neural predisposition. Stem Cells Dev 2006; 15(2): 141-64.
[http://dx.doi.org/10.1089/scd.2006.15.141] [PMID: 16646662]
[28]
Arboleda D, Forostyak S, Jendelova P, et al. Transplantation of predifferentiated adipose-derived stromal cells for the treatment of spinal cord injury. Cell Mol Neurobiol 2011; 31(7): 1113-22.
[http://dx.doi.org/10.1007/s10571-011-9712-3] [PMID: 21630007]
[29]
Zhou Z, Chen Y, Zhang H, et al. Comparison of mesenchymal stromal cells from human bone marrow and adipose tissue for the treatment of spinal cord injury. Cytotherapy 2013; 15(4): 434-48.
[http://dx.doi.org/10.1016/j.jcyt.2012.11.015] [PMID: 23376106]
[30]
Wright KT, El Masri W, Osman A, Chowdhury J, Johnson WE. Concise review: Bone marrow for the treatment of spinal cord injury: mechanisms and clinical applications. Stem Cells 2011; 29(2): 169-78.
[http://dx.doi.org/10.1002/stem.570] [PMID: 21732476]
[31]
Hawryluk GW, Mothe A, Wang J, Wang S, Tator C, Fehlings MG. 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(12): 2222-38.
[http://dx.doi.org/10.1089/scd.2011.0596] [PMID: 22085254]
[32]
Zhukareva V, Obrocka M, Houle JD, Fischer I, Neuhuber B. Secretion profile of human bone marrow stromal cells: donor variability and response to inflammatory stimuli. Cytokine 2010; 50(3): 317-21.
[http://dx.doi.org/10.1016/j.cyto.2010.01.004] [PMID: 20185331]
[33]
White SV, Czisch CE, Han MH, Plant CD, Harvey AR, Plant GW. Intravenous Transplantation of Mesenchymal Progenitors Distribute Solely to the Lungs and Improve Outcomes in Cervical Spinal Cord Injury. Stem Cells 2016; 34(7): 1812-25.
[http://dx.doi.org/10.1002/stem.2364] [PMID: 26989838]
[34]
Neuhuber B, Timothy Himes B, Shumsky JS, Gallo G, Fischer I. Axon growth and recovery of function supported by human bone marrow stromal cells in the injured spinal cord exhibit donor variations. Brain Res 2005; 1035(1): 73-85.
[http://dx.doi.org/10.1016/j.brainres.2004.11.055] [PMID: 15713279]
[35]
Sandner B, Rivera FJ, Caioni M, et al. Bone morphogenetic proteins prevent bone marrow stromal cell-mediated oligodendroglial differentiation of transplanted adult neural progenitor cells in the injured spinal cord. Stem Cell Res (Amst) 2013; 11(2): 758-71.
[http://dx.doi.org/10.1016/j.scr.2013.05.003] [PMID: 23770801]
[36]
Quertainmont R, Cantinieaux D, Botman O, Sid S, Schoenen J, Franzen R. Mesenchymal stem cell graft improves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One 2012; 7(6), e39500
[http://dx.doi.org/10.1371/journal.pone.0039500] [PMID: 22745769]
[37]
Boido M, Garbossa D, Fontanella M, Ducati A, Vercelli A. Mesenchymal stem cell transplantation reduces glial cyst and improves functional outcome after spinal cord compression. World Neurosurg 2014; 81(1): 183-90.
[http://dx.doi.org/10.1016/j.wneu.2012.08.014] [PMID: 23022648]
[38]
Penha EM, Meira CS, Guimarães ET, et al. Use of autologous mesenchymal stem cells derived from bone marrow for the treatment of naturally injured spinal cord in dogs. Stem Cells Int 2014; 2014, 437521
[http://dx.doi.org/10.1155/2014/437521] [PMID: 24723956]
[39]
Moviglia GA, Varela G, Brizuela JA, et al. Case report on the clinical results of a combined cellular therapy for chronic spinal cord injured patients. Spinal Cord 2009; 47(6): 499-503.
[http://dx.doi.org/10.1038/sc.2008.164] [PMID: 19223861]
[40]
Liu J, Han D, Wang Z, et al. Clinical analysis of the treatment of spinal cord injury with umbilical cord mesenchymal stem cells. Cytotherapy 2013; 15(2): 185-91.
[http://dx.doi.org/10.1016/j.jcyt.2012.09.005] [PMID: 23321330]
[41]
Neirinckx V, Cantinieaux D, Coste C, Rogister B, Franzen R, Wislet-Gendebien S. Concise review: Spinal cord injuries: how could adult mesenchymal and neural crest stem cells take up the challenge? Stem Cells 2014; 32(4): 829-43.
[http://dx.doi.org/10.1002/stem.1579] [PMID: 24155224]
[42]
Cantinieaux D, Quertainmont R, Blacher S, et al. Conditioned medium from bone marrow-derived mesenchymal stem cells improves recovery after spinal cord injury in rats: an original strategy to avoid cell transplantation. PLoS One 2013; 8(8), e69515
[http://dx.doi.org/10.1371/journal.pone.0069515] [PMID: 24013448]
[43]
Yong KW, Choi JR, Dolbashid AS, Wan Safwani WKZ. Biosafety and bioefficacy assessment of human mesenchymal stem cells: what do we know so far? Regen Med 2018; 13(2): 219-32.
[http://dx.doi.org/10.2217/rme-2017-0078] [PMID: 29509072]
[44]
Lane MA, Lepore AC, Fischer I. Improving the therapeutic efficacy of neural progenitor cell transplantation following spinal cord injury. Expert Rev Neurother 2017; 17(5): 433-40.
[http://dx.doi.org/10.1080/14737175.2017.1270206] [PMID: 27927055]
[45]
Lee KZ, Lane MA, Dougherty BJ, et al. Intraspinal transplantation and modulation of donor neuron electrophysiological activity. Exp Neurol 2014; 251: 47-57.
[http://dx.doi.org/10.1016/j.expneurol.2013.10.016] [PMID: 24192152]
[46]
Lu P, Jones LL, Snyder EY, Tuszynski MH. Neural stem cells constitutively secrete neurotrophic factors and promote extensive host axonal growth after spinal cord injury. Exp Neurol 2003; 181(2): 115-29.
[http://dx.doi.org/10.1016/S0014-4886(03)00037-2] [PMID: 12781986]
[47]
Bonner JF, Steward O. Repair of spinal cord injury with neuronal relays: From fetal grafts to neural stem cells. Brain Res 2015; 1619: 115-23.
[http://dx.doi.org/10.1016/j.brainres.2015.01.006] [PMID: 25591483]
[48]
Xiong LL, Zou Y, Shi Y, et al. Tree shrew neural stem cell transplantation promotes functional recovery of tree shrews with a hemi‑sectioned spinal cord injury by upregulating nerve growth factor expression. Int J Mol Med 2018; 41(6): 3267-77.
[http://dx.doi.org/10.3892/ijmm.2018.3553] [PMID: 29532893]
[49]
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): 484-90.
[http://dx.doi.org/10.1038/nm.4502] [PMID: 29480894]
[50]
Åkesson E, Sundström E. Human neural progenitor cells in central nervous system lesions. Best Pract Res Clin Obstet Gynaecol 2016; 31: 69-81.
[http://dx.doi.org/10.1016/j.bpobgyn.2015.11.020] [PMID: 26803559]
[51]
Kameda T, Imamura T, Nakashima K. Epigenetic regulation of neural stem cell differentiation towards spinal cord regeneration. Cell Tissue Res 2018; 371(1): 189-99.
[http://dx.doi.org/10.1007/s00441-017-2656-2] [PMID: 28695279]
[52]
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(9): 3255-66.
[http://dx.doi.org/10.1172/JCI42957] [PMID: 20714104]
[53]
Yokota K, Kobayakawa K, Kubota K, et al. Engrafted Neural Stem/Progenitor Cells Promote Functional Recovery through Synapse Reorganization with Spared Host Neurons after Spinal Cord Injury. Stem Cell Reports 2015; 5(2): 264-77.
[http://dx.doi.org/10.1016/j.stemcr.2015.06.004] [PMID: 26190527]
[54]
Salewski RP, Mitchell RA, Shen C, Fehlings MG. 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.
[http://dx.doi.org/10.1089/scd.2014.0096] [PMID: 25119334]
[55]
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(7): 708-19.
[http://dx.doi.org/10.5966/sctm.2014-0215] [PMID: 26019226]
[56]
Sharp J, Frame J, Siegenthaler M, Nistor G, Keirstead HS. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants improve recovery after cervical spinal cord injury. Stem Cells 2010; 28(1): 152-63.
[PMID: 19877167]
[57]
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
[http://dx.doi.org/10.1371/journal.pone.0011852] [PMID: 20686613]
[58]
Närvä E, Autio R, Rahkonen N, et al. High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced copy number changes and loss of heterozygosity. Nat Biotechnol 2010; 28(4): 371-7.
[http://dx.doi.org/10.1038/nbt.1615] [PMID: 20351689]
[59]
Johnson PJ, Tatara A, Shiu A, Sakiyama-Elbert SE. Controlled release of neurotrophin-3 and platelet-derived growth factor from fibrin scaffolds containing neural progenitor cells enhances survival and differentiation into neurons in a subacute model of SCI. Cell Transplant 2010; 19(1): 89-101.
[http://dx.doi.org/10.3727/096368909X477273] [PMID: 19818206]
[60]
Marichal N, García G, Radmilovich M, Trujillo-Cenóz O, Russo RE. Spatial domains of progenitor-like cells and functional complexity of a stem cell niche in the neonatal rat spinal cord. Stem Cells 2012; 30(9): 2020-31.
[http://dx.doi.org/10.1002/stem.1175] [PMID: 22821702]
[61]
Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126(4): 663-76.
[http://dx.doi.org/10.1016/j.cell.2006.07.024] [PMID: 16904174]
[62]
Wang H, Fang H, Dai J, Liu G, Xu ZJ. Induced pluripotent stem cells for spinal cord injury therapy: current status and perspective. Neurol Sci 2013; 34(1): 11-7.
[http://dx.doi.org/10.1007/s10072-012-1145-3] [PMID: 22797773]
[63]
Kim JB, Zaehres H, Wu G, et al. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature 2008; 454(7204): 646-50.
[http://dx.doi.org/10.1038/nature07061] [PMID: 18594515]
[64]
Liu P, Chen M, Liu Y, Qi LS, Ding S. CRISPR-Based Chromatin Remodeling of the Endogenous Oct4 or Sox2 Locus Enables Reprogramming to Pluripotency. Cell Stem Cell 2018; 22(2): 252-261.e4.
[http://dx.doi.org/10.1016/j.stem.2017.12.001] [PMID: 29358044]
[65]
Mallon BS, Hamilton RS, Kozhich OA, et al. Comparison of the molecular profiles of human embryonic and induced pluripotent stem cells of isogenic origin. Stem Cell Res (Amst) 2014; 12(2): 376-86.
[http://dx.doi.org/10.1016/j.scr.2013.11.010] [PMID: 24374290]
[66]
Hirasaki M, Hiraki-Kamon K, Kamon M, et al. Striking similarity in the gene expression levels of individual Myc module members among ESCs, EpiSCs, and partial iPSCs. PLoS One 2013; 8(12), e83769
[http://dx.doi.org/10.1371/journal.pone.0083769] [PMID: 24386274]
[67]
Chen KG, Mallon BS, McKay RD, Robey PG. Human pluripotent stem cell culture: considerations for maintenance, expansion, and therapeutics. Cell Stem Cell 2014; 14(1): 13-26.
[http://dx.doi.org/10.1016/j.stem.2013.12.005] [PMID: 24388173]
[68]
Tsuji O, Miura K, Okada Y, et al. Therapeutic potential of appropriately evaluated safe-induced pluripotent stem cells for spinal cord injury. Proc Natl Acad Sci USA 2010; 107(28): 12704-9.
[http://dx.doi.org/10.1073/pnas.0910106107] [PMID: 20615974]
[69]
Okada Y, Matsumoto A, Shimazaki T, et al. Spatiotemporal recapitulation of central nervous system development by murine embryonic stem cell-derived neural stem/progenitor cells. Stem Cells 2008; 26(12): 3086-98.
[http://dx.doi.org/10.1634/stemcells.2008-0293] [PMID: 18757299]
[70]
Nori S, Okada Y, Yasuda A, et al. Grafted human-induced pluripotent stem-cell-derived neurospheres promote motor functional recovery after spinal cord injury in mice. Proc Natl Acad Sci USA 2011; 108(40): 16825-30.
[http://dx.doi.org/10.1073/pnas.1108077108] [PMID: 21949375]
[71]
Fujimoto Y, Abematsu M, Falk A, et al. Treatment of a mouse model of spinal cord injury by transplantation of human induced pluripotent stem cell-derived long-term self-renewing neuroepithelial-like stem cells. Stem Cells 2012; 30(6): 1163-73.
[http://dx.doi.org/10.1002/stem.1083] [PMID: 22419556]
[72]
Kobayashi Y, Okada Y, Itakura G, et al. Pre-evaluated safe human iPSC-derived neural stem cells promote functional recovery after spinal cord injury in common marmoset without tumorigenicity. PLoS One 2012; 7(12), e52787
[http://dx.doi.org/10.1371/journal.pone.0052787] [PMID: 23300777]
[73]
Okano H, Nakamura M, Yoshida K, et al. Steps toward safe cell therapy using induced pluripotent stem cells. Circ Res 2013; 112(3): 523-33.
[http://dx.doi.org/10.1161/CIRCRESAHA.111.256149] [PMID: 23371901]
[74]
Itakura G, Kobayashi Y, Nishimura S, et al. Controlling immune rejection is a fail-safe system against potential tumorigenicity after human iPSC-derived neural stem cell transplantation. PLoS One 2015; 10(2), e0116413
[http://dx.doi.org/10.1371/journal.pone.0116413] [PMID: 25706286]
[75]
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 Reports 2016; 7(4): 649-63.
[http://dx.doi.org/10.1016/j.stemcr.2016.08.015] [PMID: 27666789]
[76]
Dakhore S, Nayer B, Hasegawa K. Human Pluripotent Stem Cell Culture: Current Status, Challenges, and Advancement. Stem Cells Int 2018; 2018, 7396905
[http://dx.doi.org/10.1155/2018/7396905] [PMID: 30595701]
[77]
Higuchi A, Suresh Kumar S, Benelli G, et al. Biomaterials used in stem cell therapy for spinal cord injury. Prog Mater Sci 2019; 103: 374-424.
[http://dx.doi.org/10.1016/j.pmatsci.2019.02.002]
[78]
Angeli CA, Boakye M, Morton RA, et al. Recovery of over-ground walking after chronic motor complete spinal cord injury. N Engl J Med 2018; 379(13): 1244-50.
[http://dx.doi.org/10.1056/NEJMoa1803588] [PMID: 30247091]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy