J Korean Orthop Assoc.  2019 Dec;54(6):498-508. 10.4055/jkoa.2019.54.6.498.

Spinal Cord Injury and Neuro-Regeneration

Affiliations
  • 1Department of Orthopedic Surgery, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Korea. boscoa@catholic.ac.kr

Abstract

Recent advances in the understanding of the pathophysiology of spinal cord injury (SCI) and new therapeutic approaches have provided promising results for this incurable and debilitating central nervous system injury. Various neuro-protective and neuro-regenerative trials have been attempted to overcome SCIs. This review summarizes the reported experimental and clinical data regarding neuro-regenerative trials with the proven pathophysiology of SCI. In addition, the prerequisites for safe and effective clinical trials are discussed.

Keyword

spinal cord injuries; neuroprotection; spinal cord regeneration; cell-and tissue-based therapy

MeSH Terms

Central Nervous System
Neuroprotection
Spinal Cord Injuries*
Spinal Cord Regeneration
Spinal Cord*

Figure

  • Figure 1 Pathomechanism of spinal cord injuries. Primary injury means direct mechanical injury to the spinal cord. And secondary injury means consequent injury mechanism following direct injury. Ischemia, inflammation, ionic dysfunction, excitotoxicity, and reperfusion mechanism are related to this secondary mechanism. Various trials to intervene these mechanism have been investigated to find the therapeutic strategy for spinal cord injuries.


Reference

1. Sekhon LH, Fehlings MG. Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine (Phila Pa 1976). 2001; 26:S2–12.
Article
2. Priebe MM, Chiodo AE, Scelza WM, Kirshblum SC, Wuermser LA, Ho CH. Spinal cord injury medicine. 6. Economic and societal issues in spinal cord injury. Arch Phys Med Rehabil. 2007; 88:S84–S88.
Article
3. Ho CH, Wuermser LA, Priebe MM, Chiodo AE, Scelza WM, Kirshblum SC. Spinal cord injury medicine. 1. Epidemiology and classification. Arch Phys Med Rehabil. 2007; 88:S49–S54.
Article
4. National Spinal Cord Injury Statistical Center. Spinal cord injury. Facts and figures at a glance. J Spinal Cord Med. 2005; 28:379–380.
5. Barron KD, Hirano A, Araki S, Terry RD. Experiences with metastatic neoplasms involving the spinal cord. Neurology. 1959; 9:91–106.
Article
6. Gerszten PC, Welch WC. Current surgical management of metastatic spinal disease. Oncology (Williston Park). 2000; 14:1013–1024.
7. Schaberg J, Gainor BJ. A profile of metastatic carcinoma of the spine. Spine (Phila Pa 1976). 1985; 10:19–20.
Article
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:E2.
Article
9. Fehlings MG, Tator CH. The relationships among the severity of spinal cord injury, residual neurological function, axon counts, and counts of retrogradely labeled neurons after experimental spinal cord injury. Exp Neurol. 1995; 132:220–228.
Article
10. Kakulas BA. Neuropathology: the foundation for new treatments in spinal cord injury. Spinal Cord. 2004; 42:549–563.
Article
11. Kwon BK, Tetzlaff W, Grauer JN, Beiner J, Vaccaro AR. Pathophysiology and pharmacologic treatment of acute spinal cord injury. Spine J. 2004; 4:451–464.
Article
12. 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.
Article
13. Tator CH, Koyanagi I. Vascular mechanisms in the pathophysiology of human spinal cord injury. J Neurosurg. 1997; 86:483–492.
Article
14. Ha KY, Carragee E, Cheng I, Kwon SE, Kim YH. Pregabalin as a neuroprotector after spinal cord injury in rats: biochemical analysis and effect on glial cells. J Korean Med Sci. 2011; 26:404–411.
Article
15. Ha KY, Kim YH, Rhyu KW, Kwon SE. Pregabalin as a neuroprotector after spinal cord injury in rats. Eur Spine J. 2008; 17:864–872.
Article
16. Schanne FA, Kane AB, Young EE, Farber JL. Calcium dependence of toxic cell death: a final common pathway. Science. 1979; 206:700–702.
Article
17. Li S, Stys PK. Mechanisms of ionotropic glutamate receptor-mediated excitotoxicity in isolated spinal cord white matter. J Neurosci. 2000; 20:1190–1198.
Article
18. Li S, Mealing GA, Morley P, Stys PK. Novel injury mechanism in anoxia and trauma of spinal cord white matter: glutamate release via reverse Na+-dependent glutamate transport. J Neurosci. 1999; 19:RC16.
Article
19. Park E, Velumian AA, Fehlings MG. The role of excitotoxicity in secondary mechanisms of spinal cord injury: a review with an emphasis on the implications for white matter degeneration. J Neurotrauma. 2004; 21:754–774.
Article
20. Donnelly DJ, Popovich PG. Inflammation and its role in neuroprotection, axonal regeneration and functional recovery after spinal cord injury. Exp Neurol. 2008; 209:378–388.
Article
21. Xiong Y, Rabchevsky AG, Hall ED. Role of peroxynitrite in secondary oxidative damage after spinal cord injury. J Neurochem. 2007; 100:639–649.
Article
22. Pineau I, Lacroix S. Proinflammatory cytokine synthesis in the injured mouse spinal cord: multiphasic expression pattern and identification of the cell types involved. J Comp Neurol. 2007; 500:267–285.
Article
23. Noble LJ, Wrathall JR. Distribution and time course of protein extravasation in the rat spinal cord after contusive injury. Brain Res. 1989; 482:57–66.
Article
24. Fleming JC, Norenberg MD, Ramsay DA, et al. The cellular inflammatory response in human spinal cords after injury. Brain. 2006; 129:3249–3269.
Article
25. Popovich PG, Wei P, Stokes BT. Cellular inflammatory response after spinal cord injury in Sprague-Dawley and Lewis rats. J Comp Neurol. 1997; 377:443–464.
Article
26. Bethea JR, Nagashima H, Acosta MC, et al. Systemically administered interleukin-10 reduces tumor necrosis factor-alpha production and significantly improves functional recovery following traumatic spinal cord injury in rats. J Neurotrauma. 1999; 16:851–863.
Article
27. Kim GM, Xu J, Xu J, et al. Tumor necrosis factor receptor deletion reduces nuclear factor-kappaB activation, cellular inhibitor of apoptosis protein 2 expression, and functional recovery after traumatic spinal cord injury. J Neurosci. 2001; 21:6617–6625.
28. Yan P, Li Q, Kim GM, Xu J, Hsu CY, Xu XM. Cellular localization of tumor necrosis factor-alpha following acute spinal cord injury in adult rats. J Neurotrauma. 2001; 18:563–568.
29. Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. Prog Brain Res. 2002; 137:37–47.
30. Fan LH, Wang KZ, Cheng B, Wang CS, Dang XQ. Anti-apoptotic and neuroprotective effects of Tetramethylpyrazine following spinal cord ischemia in rabbits. BMC Neurosci. 2006; 7:48.
Article
31. Lou J, Lenke LG, Ludwig FJ, O'Brien MF. Apoptosis as a mechanism of neuronal cell death following acute experimental spinal cord injury. Spinal Cord. 1998; 36:683–690.
Article
32. Ha KY, Kim YH. Neuroprotective effect of moderate epidural hypothermia after spinal cord injury in rats. Spine (Phila Pa 1976). 2008; 33:2059–2065.
Article
33. Stys PK, Lipton SA. White matter NMDA receptors: an unexpected new therapeutic target? Trends Pharmacol Sci. 2007; 28:561–566.
Article
34. Agrawal SK, Fehlings MG. Mechanisms of secondary injury to spinal cord axons in vitro: role of Na+, Na(+)-K(+)-ATPase, the Na(+)-H+ exchanger, and the Na(+)-Ca2+ exchanger. J Neurosci. 1996; 16:545–552.
Article
35. Totoiu MO, Keirstead HS. Spinal cord injury is accompanied by chronic progressive demyelination. J Comp Neurol. 2005; 486:373–383.
Article
36. Lasiene J, Shupe L, Perlmutter S, Horner P. No evidence for chronic demyelination in spared axons after spinal cord injury in a mouse. J Neurosci. 2008; 28:3887–3896.
Article
37. Anderson CM, Swanson RA. Astrocyte glutamate transport: review of properties, regulation, and physiological functions. Glia. 2000; 32:1–14.
Article
38. Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci. 2004; 24:2143–2155.
Article
39. Williams A, Piaton G, Lubetzki C. Astrocytes: friends or foes in multiple sclerosis? Glia. 2007; 55:1300–1312.
40. 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; 26:3377–3389.
Article
41. Herrmann JE, Imura T, Song B, et al. STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci. 2008; 28:7231–7243.
Article
42. Buffo A, Rolando C, Ceruti S. Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential. Biochem Pharmacol. 2010; 79:77–89.
Article
43. McDonald JW, Becker D, Sadowsky CL, Jane JA Sr, Conturo TE, Schultz LM. Late recovery following spinal cord injury. Case report and review of the literature. J Neurosurg. 2002; 97:252–265.
44. Duncan ID, Brower A, Kondo Y, Curlee JF Jr, Schultz RD. Extensive remyelination of the CNS leads to functional recovery. Proc Natl Acad Sci U S A. 2009; 106:6832–6836.
Article
45. Liebetanz D, Merkler D. Effects of commissural de- and remyelination on motor skill behaviour in the cuprizone mouse model of multiple sclerosis. Exp Neurol. 2006; 202:217–224.
Article
46. Blight AR. Delayed demyelination and macrophage invasion: a candidate for secondary cell damage in spinal cord injury. Cent Nerv Syst Trauma. 1985; 2:299–315.
Article
47. Biernaskie J, Sparling JS, Liu J, et al. Skin-derived precursors generate myelinating Schwann cells that promote remyelination and functional recovery after contusion spinal cord injury. J Neurosci. 2007; 27:9545–9559.
Article
48. Bretzner F, Plemel JR, Liu J, Richter M, Roskams AJ, Tetzlaff W. Combination of olfactory ensheathing cells with local versus systemic cAMP treatment after a cervical rubrospinal tract injury. J Neurosci Res. 2010; 88:2833–2846.
Article
49. Ramer LM, Au E, Richter MW, Liu J, Tetzlaff W, Roskams AJ. Peripheral olfactory ensheathing cells reduce scar and cavity formation and promote regeneration after spinal cord injury. J Comp Neurol. 2004; 473:1–15.
Article
50. Ramer LM, Richter MW, Roskams AJ, Tetzlaff W, Ramer MS. Peripherally-derived olfactory ensheathing cells do not promote primary afferent regeneration following dorsal root injury. Glia. 2004; 47:189–206.
Article
51. Waxman SG. Axonal conduction and injury in multiple sclerosis: the role of sodium channels. Nat Rev Neurosci. 2006; 7:932–941.
Article
52. Guest JD, Hiester ED, Bunge RP. Demyelination and Schwann cell responses adjacent to injury epicenter cavities following chronic human spinal cord injury. Exp Neurol. 2005; 192:384–393.
Article
53. Cheng H, Cao Y, Olson L. Spinal cord repair in adult paraplegic rats: partial restoration of hind limb function. Science. 1996; 273:510–513.
Article
54. 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. 2005; 25:4694–4705.
Article
55. Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS. Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia. 2005; 49:385–396.
Article
56. Horky LL, Galimi F, Gage FH, Horner PJ. Fate of endogenous stem/progenitor cells following spinal cord injury. J Comp Neurol. 2006; 498:525–538.
Article
57. Lytle JM, Chittajallu R, Wrathall JR, Gallo V. NG2 cell response in the CNP-EGFP mouse after contusive spinal cord injury. Glia. 2009; 57:270–285.
Article
58. Barres BA, Schmid R, Sendnter M, Raff MC. Multiple extracellular signals are required for long-term oligodendrocyte survival. Development. 1993; 118:283–295.
Article
59. Tripathi RB, McTigue DM. Chronically increased ciliary neurotrophic factor and fibroblast growth factor-2 expression after spinal contusion in rats. J Comp Neurol. 2008; 510:129–144.
Article
60. Caroni P, Schwab ME. Antibody against myelin-associated inhibitor of neurite growth neutralizes nonpermissive substrate properties of CNS white matter. Neuron. 1988; 1:85–96.
61. Caroni P, Schwab ME. Two membrane protein fractions from rat central myelin with inhibitory properties for neurite growth and fibroblast spreading. J Cell Biol. 1988; 106:1281–1288.
Article
62. Monnier PP, Sierra A, Schwab JM, Henke-Fahle S, Mueller BK. The Rho/ROCK pathway mediates neurite growth-inhibitory activity associated with the chondroitin sulfate proteoglycans of the CNS glial scar. Mol Cell Neurosci. 2003; 22:319–330.
Article
63. Madura T, Yamashita T, Kubo T, Fujitani M, Hosokawa K, Tohyama M. Activation of Rho in the injured axons following spinal cord injury. EMBO Rep. 2004; 5:412–417.
Article
64. Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 2005; 4:387–398.
Article
65. Borisoff JF, Chan CC, Hiebert GW, et al. Suppression of Rho-kinase activity promotes axonal growth on inhibitory CNS substrates. Mol Cell Neurosci. 2003; 22:405–416.
Article
66. Dergham P, Ellezam B, Essagian C, Avedissian H, Lubell WD, McKerracher L. Rho signaling pathway targeted to promote spinal cord repair. J Neurosci. 2002; 22:6570–6577.
Article
67. Benzel EC. Spine surgery: techniques, complication avoidance, and management. 3rd ed. Philadelphia: Elsevier/Saunders;2012.
68. Barritt AW, Davies M, Marchand F, et al. Chondroitinase ABC promotes sprouting of intact and injured spinal systems after spinal cord injury. J Neurosci. 2006; 26:10856–10867.
Article
69. Bradbury EJ, Moon LD, Popat RJ, et al. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature. 2002; 416:636–640.
Article
70. Caggiano AO, Zimber MP, Ganguly A, Blight AR, Gruskin EA. Chondroitinase ABCI improves locomotion and bladder function following contusion injury of the rat spinal cord. J Neurotrauma. 2005; 22:226–239.
Article
71. Fouad K, Schnell L, Bunge MB, Schwab ME, Liebscher T, Pearse DD. Combining Schwann cell bridges and olfactory-ensheathing glia grafts with chondroitinase promotes locomotor recovery after complete transection of the spinal cord. J Neurosci. 2005; 25:1169–1178.
Article
72. Shields LB, Zhang YP, Burke DA, Gray R, Shields CB. Benefit of chondroitinase ABC on sensory axon regeneration in a laceration model of spinal cord injury in the rat. Surg Neurol. 2008; 69:568–577.
Article
73. Kim JW, Ha KY, Molon JN, Kim YH. Bone marrow-derived mesenchymal stem cell transplantation for chronic spinal cord injury in rats: comparative study between intralesional and intravenous transplantation. Spine (Phila Pa 1976). 2013; 38:E1065–E1074.
74. Nishimura S, Yasuda A, Iwai H, et al. Time-dependent changes in the microenvironment of injured spinal cord affects the therapeutic potential of neural stem cell transplantation for spinal cord injury. Mol Brain. 2013; 6:3.
Article
75. Crigler L, Robey RC, Asawachaicharn A, Gaupp D, Phinney DG. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp Neurol. 2006; 198:54–64.
Article
76. 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; 27:307–321.
Article
77. Bartholomew A, Sturgeon C, Siatskas M, et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol. 2002; 30:42–48.
Article
78. Nakajima H, Uchida K, Guerrero AR, et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J Neurotrauma. 2012; 29:1614–1625.
Article
79. 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; 21:2222–2238.
Article
80. Dasari VR, Veeravalli KK, Tsung AJ, et al. Neuronal apoptosis is inhibited by cord blood stem cells after spinal cord injury. J Neurotrauma. 2009; 26:2057–2069.
Article
81. Dasari VR, Spomar DG, Li L, Gujrati M, Rao JS, Dinh DH. Umbilical cord blood stem cell mediated downregulation of fas improves functional recovery of rats after spinal cord injury. Neurochem Res. 2008; 33:134–149.
Article
82. 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.
Article
83. Kim DH, Yoo KH, Yim YS, et al. Cotransplanted bone marrow derived mesenchymal stem cells (MSC) enhanced engraftment of hematopoietic stem cells in a MSC-dose dependent manner in NOD/SCID mice. J Korean Med Sci. 2006; 21:1000–1004.
Article
84. Ikegami T, Nakamura M, Yamane J, et al. Chondroitinase ABC combined with neural stem/progenitor cell transplantation enhances graft cell migration and outgrowth of growth-associated protein-43-positive fibers after rat spinal cord injury. Eur J Neurosci. 2005; 22:3036–3046.
Article
85. Massey JM, Amps J, Viapiano MS, et al. Increased chondroitin sulfate proteoglycan expression in denervated brainstem targets following spinal cord injury creates a barrier to axonal regeneration overcome by chondroitinase ABC and neurotrophin-3. Exp Neurol. 2008; 209:426–445.
Article
86. Cafferty WB, Bradbury EJ, Lidierth M, et al. Chondroitinase ABC-mediated plasticity of spinal sensory function. J Neurosci. 2008; 28:11998–12009.
Article
87. 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:e85916.
Article
88. Lee HJ, Wu J, Chung J, Wrathall JR. SOX2 expression is upregulated in adult spinal cord after contusion injury in both oligodendrocyte lineage and ependymal cells. J Neurosci Res. 2013; 91:196–210.
Article
89. Panayiotou E, Malas S. Adult spinal cord ependymal layer: a promising pool of quiescent stem cells to treat spinal cord injury. Front Physiol. 2013; 4:340.
Article
90. Lee JY, Ha KY, Kim JW, Seo JY, Kim YH. Does extracorporeal shock wave introduce alteration of microenvironment in cell therapy for chronic spinal cord injury? Spine (Phila Pa 1976). 2014; 39:E1553–E1559.
Article
91. Shin DC, Ha KY, Kim YH, Kim JW, Cho YK, Kim SI. Induction of endogenous neural stem cells by extracorporeal shock waves after spinal cord injury. Spine (Phila Pa 1976). 2018; 43:E200–E207.
Article
92. 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.
Article
Full Text Links
  • JKOA
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr