Journal Home > Volume 5 , Issue 1

Spinal cord injury (SCI) is a devastating condition and major burden on society and individuals. Currently, neurorestorative strategies, including stem cell therapy products or mature/functionally differentiated cell-derived cell therapy products, can restore patients with chronic complete SCI to some degree of neurological functions. The stem cells for neurorestoration include neural stem cells, mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, etc. A better understanding of the merits, demerits and precise function of different stem cells in the treatment of SCI may aid in the development of neurorestorative strategies. However, the efficacy, safety and ethical concerns of stem cell-based therapy continue to be challenged. Nonetheless, stem cell-based therapies hold promise of widespread applications, particularly in areas of SCI, and have the potential to be novel therapeutics, which contributes to the repair of SCI. This review mainly focused on recent advances regarding the stem cell-based therapies in the treatment of SCI and discussed future perspectives in this field.


menu
Abstract
Full text
Outline
About this article

Stem cell-based therapies to treat spinal cord injury: a review

Show Author's information Zhongju Shi1,2Hongyun Huang3Shiqing Feng1,2( )
Department of Orthopaedics, Tianjin Medical University General Hospital,
Institute of Neurology, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin City, Tianjin, 3Institute of Neurorestoratology, General Hospital of Armed Police Forces, Beijing, People’s Republic of China

Abstract

Spinal cord injury (SCI) is a devastating condition and major burden on society and individuals. Currently, neurorestorative strategies, including stem cell therapy products or mature/functionally differentiated cell-derived cell therapy products, can restore patients with chronic complete SCI to some degree of neurological functions. The stem cells for neurorestoration include neural stem cells, mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, etc. A better understanding of the merits, demerits and precise function of different stem cells in the treatment of SCI may aid in the development of neurorestorative strategies. However, the efficacy, safety and ethical concerns of stem cell-based therapy continue to be challenged. Nonetheless, stem cell-based therapies hold promise of widespread applications, particularly in areas of SCI, and have the potential to be novel therapeutics, which contributes to the repair of SCI. This review mainly focused on recent advances regarding the stem cell-based therapies in the treatment of SCI and discussed future perspectives in this field.

Keywords: spinal cord injury, induced pluripotent stem cells, neural stem cells, bone marrow-derived mesenchymal stem cells, adipose-derived stem cells, embryonic stem cells

References(85)

1.
Antonic A, Sena ES, Lees JS, et al. Stem cell transplantation in traumatic spinal cord injury: a systematic review and meta-analysis of animal studies. PLoS Biol. 2013;11(12):e1001738.
2.
DeVivo MJ, Krause JS, Lammertse DP. Recent trends in mortality and causes of death among persons with spinal cord injury. Arch Phys Med Rehabil. 1999;80(11):1411–1419.
3.
Dolbow DR. Exercise following spinal cord injury: physiology to therapy. J Neurorestoratol. 2015;3:133–139.
4.
Huang H, Sun T, Chen L, et al. Consensus of clinical neurorestorative progress in patients with complete chronic spinal cord injury. Cell Transplant. 2014;23(suppl 1):S5–S17.
5.
Freeman SN, Burke KA, Imoisili MA, Cote TR. The Orphan Drug Act and the development of stem cell-based products for rare diseases. Cell Stem Cell. 2010;7(3):283–287.
6.
Doulames VM, Plant GW. Induced pluripotent stem cell therapies for cervical spinal cord injury. Int J Mol Sci. 2016;17(4):530.
7.
Kwon BK, Tetzlaff W, Grauer JN, Beiner J, Vaccaro AR. Pathophysiology and pharmacologic treatment of acute spinal cord injury. Spine J. 2004;4(4):451–464.
8.
Ben-Hur T. Reconstructing neural circuits using transplanted neural stem cells in the injured spinal cord. J Clin Invest. 2010;120(9):3096–3098.
9.
McDonald JW, Becker D. Spinal cord injury: promising interventions and realistic goals. Am J Phys Med Rehabil. 2003;82(10 suppl):S38–S49.
10.
Ronaghi M, Erceg S, Moreno-Manzano V, Stojkovic M. Challenges of stem cell therapy for spinal cord injury: human embryonic stem cells, endogenous neural stem cells, or induced pluripotent stem cells? Stem Cells. 2010;28(1):93–99.
11.
Thomas ED, Lochte HL Jr, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957;257(11):491–496.
12.
Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology. 2013;33(5):491–504.
13.
Batista CE, Mariano ED, Marie SK, et al. Stem cells in neurology – current perspectives. Arq Neuropsiquiatr. 2014;72(6):457–465.
14.
Saberi H, Derakhshanrad N, Yekaninejad MS. Review of recently documented clinical neuroprotective and cellular treatment for spinal cord injury: an analysis of outcomes. J Neurorestoratol. 2014;2:15–24.
15.
Sandner B, Prang P, Rivera FJ, Aigner L, Blesch A, Weidner N. Neural stem cells for spinal cord repair. Cell Tissue Res. 2012;349(1):349–362.
16.
Dell’Anno MT, Strittmatter SM. Rewiring the spinal cord: direct and indirect strategies. Neurosci Lett. 2017;652:25–34.
17.
Kabatas S, Teng YD. Potential roles of the neural stem cell in the restoration of the injured spinal cord: review of the literature. Turk Neurosurg. 2010;20(2):103–110.
18.
Mothe AJ, Tator CH. Advances in stem cell therapy for spinal cord injury. J Clin Invest. 2012;122(11):3824–3834.
19.
Pfeifer K, Vroemen M, Caioni M, Aigner L, Bogdahn U, Weidner N. Autologous adult rodent neural progenitor cell transplantation represents a feasible strategy to promote structural repair in the chronically injured spinal cord. Regen Med. 2006;1(2):255–266.
20.
Shihabuddin LS, Horner PJ, Ray J, Gage FH. Adult spinal cord stem cells generate neurons after transplantation in the adult dentate gyrus. J Neurosci. 2000;20(23):8727–8735.
21.
Han SS, Kang DY, Mujtaba T, Rao MS, Fischer I. Grafted lineage-restricted precursors differentiate exclusively into neurons in the adult spinal cord. Exp Neurol. 2002;177(2):360–375.
22.
McDonald JW, Liu XZ, Qu Y, et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat Med. 1999;5(12):1410–1412.
23.
Pfeifer K, Vroemen M, Blesch A, Weidner N. Adult neural progenitor cells provide a permissive guiding substrate for corticospinal axon growth following spinal cord injury. Eur J Neurosci. 2004;20(7):1695–1704.
24.
Tuszynski MH, Steward O. Concepts and methods for the study of axonal regeneration in the CNS. Neuron. 2012;74(5):777–791.
25.
Sharp KG, Yee KM, Steward O. A re-assessment of long distance growth and connectivity of neural stem cells after severe spinal cord injury. Exp Neurol. 2014;257:186–204.
26.
Basso DM, Beattie MS, Bresnahan JC. Graded histological and locomotor outcomes after spinal cord contusion using the NYU weight-drop device versus transection. Exp Neurol. 1996;139(2):244–256.
27.
Gledhill RF, Harrison BM, McDonald WI. Demyelination and remyelination after acute spinal cord compression. Exp Neurol. 1973;38(3):472–487.
28.
Crowe MJ, Bresnahan JC, Shuman SL, Masters JN, Beattie MS. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat Med. 1997;3(1):73–76.
29.
Karimi-Abdolrezaee S, Eftekharpour E, Wang J, Morshead CM, Fehlings MG. Delayed transplantation of adult neural precursor cells promotes remyelination and functional neurological recovery after spinal cord injury. J Neurosci. 2006;26(13):3377–3389.
30.
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Myelin damage and repair in pathologic CNS: challenges and prospects. Front Mol Neurosci. 2015;8:35.
31.
Eftekharpour E, Karimi-Abdolrezaee S, Fehlings MG. Current status of experimental cell replacement approaches to spinal cord injury. Neurosurg Focus. 2008;24(3–4):E19.
32.
Cummings BJ, Uchida N, Tamaki SJ, et al. Human neural stem cells differentiate and promote locomotor recovery in spinal cord-injured mice. Proc Natl Acad Sci U S A. 2005;102(39):14069–14074.
33.
Chopp M, Zhang XH, Li Y, et al. Spinal cord injury in rat: treatment with bone marrow stromal cell transplantation. Neuroreport. 2000;11(13):3001–3005.
34.
Zurita M, Vaquero J. Bone marrow stromal cells can achieve cure of chronic paraplegic rats: functional and morphological outcome one year after transplantation. Neurosci Lett. 2006;402(1–2):51–56.
35.
Wu S, Suzuki Y, Ejiri Y, et al. Bone marrow stromal cells enhance differentiation of cocultured neurosphere cells and promote regeneration of injured spinal cord. J Neurosci Res. 2003;72(3):343–351.
36.
Li G, Che MT, Zhang K, et al. Graft of the NT-3 persistent delivery gelatin sponge scaffold promotes axon regeneration, attenuates inflammation, and induces cell migration in rat and canine with spinal cord injury. Biomaterials. 2016;83:233–248.
37.
Abrams MB, Dominguez C, Pernold K, et al. Multipotent mesenchymal stromal cells attenuate chronic inflammation and injury-induced sensitivity to mechanical stimuli in experimental spinal cord injury. Restor Neurol Neurosci. 2009;27(4):307–321.
38.
Karamouzian S, Nematollahi-Mahani SN, Nakhaee N, Eskandary H. Clinical safety and primary efficacy of bone marrow mesenchymal cell transplantation in subacute spinal cord injured patients. Clin Neurol Neurosurg. 2012;114(7):935–939.
39.
Kumagai G, Tsoulfas P, Toh S, McNiece I, Bramlett HM, Dietrich WD. Genetically modified mesenchymal stem cells (MSCs) promote axonal regeneration and prevent hypersensitivity after spinal cord injury. Exp Neurol. 2013;248:369–380.
40.
Woodbury D, Schwarz EJ, Prockop DJ, Black IB. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res. 2000;61(4):364–370.
DOI
41.
Zhao H, Cheng L, Du X, et al. Transplantation of cerebral dopamine neurotrophic factor transducted BMSCs in contusion spinal cord injury of rats: promotion of nerve regeneration by alleviating neuroinflammation. Mol Neurobiol. 2016;53(1):187–199.
42.
Barrachina L, Remacha AR, Romero A, et al. Effect of inflammatory environment on equine bone marrow derived mesenchymal stem cells immunogenicity and immunomodulatory properties. Vet Immunol Immunopathol. 2016;171:57–65.
43.
Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death Differ. 2014;21(2):216–225.
44.
Iyer SS, Torres-Gonzalez E, Neujahr DC, et al. Effect of bone marrow-derived mesenchymal stem cells on endotoxin-induced oxidation of plasma cysteine and glutathione in mice. Stem Cells Int. 2010;2010:868076.
45.
Urdzikova L, Jendelova P, Glogarova K, Burian M, Hajek M, Sykova E. Transplantation of bone marrow stem cells as well as mobilization by granulocyte-colony stimulating factor promotes recovery after spinal cord injury in rats. J Neurotrauma. 2006;23(9):1379–1391.
46.
Akiyama Y, Radtke C, Honmou O, Kocsis JD. Remyelination of the spinal cord following intravenous delivery of bone marrow cells. Glia. 2002;39(3):229–236.
47.
Osaka M, Honmou O, Murakami T, et al. Intravenous administration of mesenchymal stem cells derived from bone marrow after contusive spinal cord injury improves functional outcome. Brain Res. 2010;1343:226–235.
48.
Vanecek V, Zablotskii V, Forostyak S, et al. Highly efficient magnetic targeting of mesenchymal stem cells in spinal cord injury. Int J Nanomedicine. 2012;7:3719–3730.
49.
Zhang RP, Wang LJ, He S, Xie J, Li JD. Effects of magnetically guided, SPIO-labeled, and neurotrophin-3 gene-modified bone mesenchymal stem cells in a rat model of spinal cord injury. Stem Cells Int. 2016;2016:2018474.
50.
Gimble JM, Katz AJ, Bunnell BA. Adipose-derived stem cells for regenerative medicine. Circ Res. 2007;100(9):1249–1260.
51.
Lin G, Garcia M, Ning H, et al. Defining stem and progenitor cells within adipose tissue. Stem Cells Dev. 2008;17(6):1053–1063.
52.
Schaffler A, Buchler C. Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells. 2007;25(4):818–827.
53.
Nogradi A, Pajer K, Marton G. The role of embryonic motoneuron transplants to restore the lost motor function of the injured spinal cord. Ann Anat. 2011;193(4):362–370.
54.
Wei Y, Gong K, Zheng Z, et al. Schwann-like cell differentiation of rat adipose-derived stem cells by indirect co-culture with Schwann cells in vitro. Cell Prolif. 2010;43(6):606–616.
55.
Gao S, Zhao P, Lin C, et al. Differentiation of human adipose-derived stem cells into neuron-like cells which are compatible with photocurable three-dimensional scaffolds. Tissue Eng Part A. 2014;20(7–8):1271–1284.
56.
Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 2001;7(2):211–228.
57.
Abdanipour A, Tiraihi T, Taheri T. 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(10):1003–1013.
58.
Kolar MK, Kingham PJ, Novikova LN, Wiberg M, Novikov LN. The therapeutic effects of human adipose-derived stem cells in a rat cervical spinal cord injury model. Stem Cells Dev. 2014;23(14):1659–1674.
59.
Oh JS, Park IS, Kim KN, Yoon DH, Kim SH, Ha Y. Transplantation of an adipose stem cell cluster in a spinal cord injury. Neuroreport. 2012;23(5):277–282.
60.
Tang L, Lu X, Zhu R, et al. Adipose-derived stem cells expressing the neurogenin-2 promote functional recovery after spinal cord injury in rat. Cell Mol Neurobiol. 2016;36(5):657–667.
61.
Schroeder GD, Kepler CK, Vaccaro AR. The use of cell transplantation in spinal cord injuries. J Am Acad Orthop Surg. 2016;24(4):266–275.
62.
Nakagawa M, Koyanagi M, Tanabe K, et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol. 2008;26(1):101–106.
63.
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. 2010;28(9):1541–1549.
64.
Plemel JR, Keough MB, Duncan GJ, et al. Remyelination after spinal cord injury: is it a target for repair? Prog Neurobiol. 2014;117:54–72.
65.
Liu S, Qu Y, Stewart TJ, et al. Embryonic stem cells differentiate into oligodendrocytes and myelinate in culture and after spinal cord transplantation. Proc Natl Acad Sci U S A. 2000;97(11):6126–6131.
66.
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):1787–1792.
67.
All AH, Gharibani P, Gupta S, et al. Early intervention for spinal cord injury with human induced pluripotent stem cells oligodendrocyte progenitors. PLoS One. 2015;10(1):e0116933.
68.
Fu X. The immunogenicity of cells derived from induced pluripotent stem cells. Cell Mol Immunol. 2014;11(1):14–16.
69.
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 U S A. 2010;107(28):12704–12709.
70.
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.
71.
Okubo T, Iwanami A, Kohyama J, et al. Pretreatment with a gamma-secretase inhibitor prevents tumor-like overgrowth in human iPSC-derived transplants for spinal cord injury. Stem Cell Reports. 2016;7(4):649–663.
72.
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):1163–1173.
73.
Romanyuk N, Amemori T, Turnovcova K, et al. Beneficial effect of human induced pluripotent stem cell-derived neural precursors in spinal cord injury repair. Cell Transplant. 2015;24(9):1781–1797.
74.
Enomoto M. Therapeutic effects of neurotrophic factors in experimental spinal cord injury models. J Neurorestoratol. 2016;4:15–22.
75.
Fuhrmann T, Anandakumaran PN, Shoichet MS. Combinatorial therapies after spinal cord injury: how can biomaterials help? Adv Healthc Mater. Epub 2017 Mar 1.
76.
Raspa A, Pugliese R, Maleki M, Gelain F. Recent therapeutic approaches for spinal cord injury. Biotechnol Bioeng. 2016;113(2):253–259.
77.
Slotkin JR, Pritchard CD, Luque B, et al. Biodegradable scaffolds promote tissue remodeling and functional improvement in non-human primates with acute spinal cord injury. Biomaterials. 2017;123:63–76.
78.
Li X, Zhao Y, Cheng S, et al. Cetuximab modified collagen scaffold directs neurogenesis of injury-activated endogenous neural stem cells for acute spinal cord injury repair. Biomaterials. 2017;137:73–86.
79.
Iyer NR, Wilems TS, Sakiyama-Elbert SE. Stem cells for spinal cord injury: strategies to inform differentiation and transplantation. Biotechnol Bioeng. 2017;114(2):245–259.
80.
Chen C, Zhao ML, Zhang RK, et al. Collagen/heparin sulfate scaffolds fabricated by a 3D bioprinter improved mechanical properties and neurological function after spinal cord injury in rats. J Biomed Mater Res A. 2017;105(5):1324–1332.
81.
Nejati-Koshki K, Mortazavi Y, Pilehvar-Soltanahmadi Y, Sheoran S, Zarghami N. An update on application of nanotechnology and stem cells in spinal cord injury regeneration. Biomed Pharmacother. 2017;90:85–92.
82.
Nagoshi N, Okano H. Applications of induced pluripotent stem cell technologies in spinal cord injury. J Neurochem. Epub 2017 Feb 15.
83.
Karimi-Abdolrezaee S, Schut D, Wang J, Fehlings MG. Chondroitinase and growth factors enhance activation and oligodendrocyte differentiation of endogenous neural precursor cells after spinal cord injury. PLoS One. 2012;7(5):e37589.
84.
Kaneko S, Iwanami A, Nakamura M, et al. A selective Sema3A inhibitor enhances regenerative responses and functional recovery of the injured spinal cord. Nat Med. 2006;12(12):1380–1389.
85.
US Food & Drug Administration. Vaccines, Blood & Biologics. Available from: https://www.fda.gov/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/default.htm. Accessed April 18, 2017.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Published: 03 July 2017
Issue date: December 2017

Copyright

© 2017 The Author(s).

Acknowledgements

This work was supported by the State Key Program of the National Natural Science Foundation of China (81330042); the International Cooperation Program of National Natural Science Foundation of China (81620108018); the Special Program for Sino-Russian Joint Research Sponsored by the Ministry of Science and Technology, China (2014DFR31210) and the Key Program Sponsored by the Tianjin Science and Technology Committee, China (13RCGFSY19000 and 14ZCZDSY00044).

Rights and permissions

© 2017 Shi et al. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).

Return