AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.1 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review | Open Access

Progression in translational research on spinal cord injury based on microenvironment imbalance

Baoyou Fan1,Zhijian Wei1,Shiqing Feng1,2 ( )
Department of Orthopedics, Tianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, 154 Anshan Road, Heping District, Tianjin 300052, China
Department of Orthopaedics, Qilu Hospital, Shandong University Centre for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China

These authors contributed equally: Baoyou Fan, Zhijian Wei

Show Author Information

Abstract

Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.

References

1

Stover, S. L., DeVivo, M. J. & Go, B. K. History, implementation, and current status of the National Spinal Cord Injury Database. Arch. Phys. Med. Rehabil. 80, 1365–1371 (1999).

2

Parent, S., Barchi, S., LeBreton, M., Casha, S. & Fehlings, M. G. The impact of specialized centers of care for spinal cord injury on length of stay, complications, and mortality: a systematic review of the literature. J. Neurotrauma 28, 1363–1370 (2011).

3

Injury, G. B. D. T. B. & Spinal Cord Injury, C. Global, regional, and national burden of traumatic brain injury and spinal cord injury, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 18, 56–87 (2019).

4

Organization, W. H. International perspectives on spinal cord injury. Weed Res. 11, 314–316 (2013).

5

Kang, Y. et al. Epidemiology of worldwide spinal cord injury: a literature review. J. Neurorestoratol. 6, 1–9 (2017).

6

Li, H. L. et al. Epidemiology of traumatic spinal cord injury in Tianjin, China: An 18-year retrospective study of 735 cases. J. Spinal Cord Med. 42, 778–785 (2019).

7

Chan, B. C., Cadarette, S. M., Wodchis, W. P., Krahn, M. D. & Mittmann, N. The lifetime cost of spinal cord injury in Ontario, Canada: A population-based study from the perspective of the public health care payer. J. Spinal Cord Med. 42, 184–193 (2019).

8

Stillman, M. D., Barber, J., Burns, S., Williams, S. & Hoffman, J. M. Complications of Spinal Cord Injury Over the First Year After Discharge From Inpatient Rehabilitation. Arch. Phys. Med. Rehabil. 98, 1800–1805 (2017).

9

LaPlaca, M. C., Simon, C. M., Prado, G. R. & Cullen, D. K. CNS injury biomechanics and experimental models. Prog. Brain Res. 161, 13–26 (2007).

10

Choo, A. M. et al. Contusion, dislocation, and distraction: primary hemorrhage and membrane permeability in distinct mechanisms of spinal cord injury. J. Neurosurg. Spine 6, 255–266 (2007).

11

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. 500, 267–285 (2007).

12

Palandi, J., Bobinski, F., de Oliveira, G. M. & Ilha, J. Neuropathic pain after spinal cord injury and physical exercise in animal models: a systematic review and meta-analysis. Neurosci. Biobehav Rev. 108, 781–795 (2020).

13
Gruner, J. A. A monitored contusion model of spinal cord injury in the rat. J. Neurotrauma 9, 123–126 (1992). discussion 126–128.
14

Stokes, B. T. Experimental spinal cord injury: a dynamic and verifiable injury device. J. Neurotrauma 9, 129–131 (1992).

15

Scheff, S. W., Rabchevsky, A. G., Fugaccia, I., Main, J. A. & Lumpp, J. E. Jr. Experimental modeling of spinal cord injury: characterization of a force-defined injury device. J. Neurotrauma 20, 179–193 (2003).

16

Marcol, W. et al. Air gun impactor−a novel model of graded white matter spinal cord injury in rodents. J. Reconstr. Microsurg. 28, 561–568 (2012).

17

Alizadeh, A., Dyck, S. M. & Karimi-Abdolrezaee, S. Traumatic Spinal Cord Injury: an overview of pathophysiology, models and acute injury mechanisms. Front. Neurol. 10, 282 (2019).

18

Rivlin, A. S. & Tator, C. H. Effect of duration of acute spinal cord compression in a new acute cord injury model in the rat. Surg. Neurol. 10, 38–43 (1978).

19

Borgens, R. B. & Shi, R. Immediate recovery from spinal cord injury through molecular repair of nerve membranes with polyethylene glycol. FASEB J. 14, 27–35 (2000).

20

Vanický, I., Urdzíková, L., Saganová, K., Cízková, D. & Gálik, J. A simple and reproducible model of spinal cord injury induced by epidural balloon inflation in the rat. J. Neurotrauma 18, 1399–1407 (2001).

21

Guha, A., Tator, C. H., Endrenyi, L. & Piper, I. Decompression of the spinal cord improves recovery after acute experimental spinal cord compression injury. Paraplegia 25, 324–339 (1987).

22

Etz, D. C. et al. Spinal cord ischemia in open and endovascular thoracoabdominal aortic aneurysm repair: new concepts. J. Cardiovasc Surg. 55, 159–168 (2014).

23

Wynn, M. M. & Acher, C. W. A modern theory of spinal cord ischemia/injury in thoracoabdominal aortic surgery and its implications for prevention of paralysis. J. Cardiothorac. Vasc. Anesth. 28, 1088–1099 (2014).

24

Tarlov, I. M. & Keener, E. B. Subarachnoid hemorrhage and tumor implants from spinal sarcoma in an infant. Neurology 3, 384–390 (1953).

25

Saadoun, S., Bell, B. A., Verkman, A. S. & Papadopoulos, M. C. Greatly improved neurological outcome after spinal cord compression injury in AQP4-deficient mice. Brain 131, 1087–1098 (2008).

26

Rosenzweig, E. S. & McDonald, J. W. Rodent models for treatment of spinal cord injury: research trends and progress toward useful repair. Curr. Opin. Neurol. 17, 121–131 (2004).

27

Li, X. et al. Scaffold-facilitated locomotor improvement post complete spinal cord injury: Motor axon regeneration versus endogenous neuronal relay formation. Biomaterials 197, 20–31 (2019).

28

Li, J. & Lepski, G. Cell transplantation for spinal cord injury: a systematic review. Biomed. Res. Int. 2013, 786475 (2013).

29

Pastrana, E. et al. Genes associated with adult axon regeneration promoted by olfactory ensheathing cells: a new role for matrix metalloproteinase 2. J. Neurosci. 26, 5347–5359 (2006).

30

Brown, A. R. & Martinez, M. Thoracic spinal cord hemisection surgery and open-field locomotor assessment in the rat. J. Vis. Exp. https://doi.org/10.3791/59738 (2019).

31

Zörner, B. et al. Profiling locomotor recovery: comprehensive quantification of impairments after CNS damage in rodents. Nat. Methods 7, 701–708 (2010).

32

Tran, A. P., Warren, P. M. & Silver, J. The biology of regeneration failure and success after spinal cord injury. Physiol. Rev. 98, 881–917 (2018).

33

Li, Y. et al. Microglia-organized scar-free spinal cord repair in neonatal mice. Nature 587, 613–618 (2020).

34

Tator, C. H. Update on the pathophysiology and pathology of acute spinal cord injury. Brain Pathol. 5, 407–413 (1995).

35

Rowland, J. W., Hawryluk, G. W., Kwon, B. & Fehlings, M. G. Current status of acute spinal cord injury pathophysiology and emerging therapies: promise on the horizon. Neurosurg. Focus 25, E2 (2008).

36

Hassannejad, Z. et al. The fate of neurons after traumatic spinal cord injury in rats: A systematic review. Iran. J. Basic Med. Sci. 21, 546–557 (2018).

37

Springer, J., Azbill, R. & Knapp, P. Activation of the caspase-3 apoptotic cascade in traumatic spinal cord injury. Nat. Med. 5, 943–946 (1999).

38

Choo, A., Liu, J., Dvorak, M., Tetzlaff, W. & Oxland, T. Secondary pathology following contusion, dislocation, and distraction spinal cord injuries. Exp. Neurol. 212, 490–506 (2008).

39

Liu, X. Z. et al. Neuronal and glial apoptosis after traumatic spinal cord injury. J. Neurosci. 17, 5395–5406 (1997).

40

Liu, M. et al. Necroptosis, a novel type of programmed cell death, contributes to early neural cells damage after spinal cord injury in adult mice. J. Spinal Cord. Med. 38, 745–753 (2015).

41

Levy, J. M. M., Towers, C. G. & Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 17, 528–542 (2017).

42

Zhang, Q., Huang, C., Meng, B., Tang, T. S. & Yang, H. L. Changes in autophagy proteins in a rat model of spinal cord injury. Chin. J. Traumatol. 17, 193–197 (2014).

43

Hernandez, D. et al. Regulation of presynaptic neurotransmission by macroautophagy. Neuron 74, 277–284 (2012).

44

Tran, A. P., Warren, P. M. & Silver, J. Regulation of autophagy by inhibitory CSPG interactions with receptor PTPsigma and its impact on plasticity and regeneration after spinal cord injury. Exp. Neurol. 328, 113276 (2020).

45

Ko, S. H., Apple, E. C., Liu, Z. & Chen, L. Age-dependent autophagy induction after injury promotes axon regeneration by limiting NOTCH. Autophagy 16, 2052–2068 (2020).

46

Hwang, J. Y., Yan, J. & Zukin, R. S. Autophagy and synaptic plasticity: epigenetic regulation. Curr. Opin. Neurobiol. 59, 207–212 (2019).

47

Ray, S. K. Modulation of autophagy for neuroprotection and functional recovery in traumatic spinal cord injury. Neural Regen. Res. 15, 1601–1612 (2020).

48

Lipinski, M. M., Wu, J., Faden, A. I. & Sarkar, C. Function and mechanisms of autophagy in brain and spinal cord trauma. Antioxid. Redox Signal. 23, 565–577 (2015).

49

Bisicchia, E. et al. Autophagy inhibition favors survival of rubrospinal neurons after spinal cord hemisection. Mol. Neurobiol. 54, 4896–4907 (2017).

50

Lin, C. W., Chen, B., Huang, K. L., Dai, Y. S. & Teng, H. L. Inhibition of autophagy by estradiol promotes locomotor recovery after spinal cord injury in rats. Neurosci. Bull. 32, 137–144 (2016).

51

Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

52

Yao, X. et al. Deferoxamine promotes recovery of traumatic spinal cord injury by inhibiting ferroptosis. Neural Regen. Res. 14, 532–541 (2019).

53

Zhang, Y. et al. Ferroptosis inhibitor SRS 16-86 attenuates ferroptosis and promotes functional recovery in contusion spinal cord injury. Brain Res. 1706, 48–57 (2019).

54

Fink, S. L. & Cookson, B. T. Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages. Cell Microbiol. 8, 1812–1825 (2006).

55

Zheng, G. et al. Carbon monoxide releasing molecule-3 alleviates neuron death after spinal cord injury via inflammasome regulation. EBioMedicine 40, 643–654 (2019).

56

Hassannejad, Z. et al. Axonal degeneration and demyelination following traumatic spinal cord injury: a systematic review and meta-analysis. J. Chem. Neuroanat. 97, 9–22 (2019).

57

Liu, K. et al. PTEN deletion enhances the regenerative ability of adult corticospinal neurons. Nat. Neurosci. 13, 1075–1081 (2010).

58

Park, K. K. et al. Promoting axon regeneration in the adult CNS by modulation of the PTEN/mTOR pathway. Science 322, 963–966 (2008).

59

Huang, H. et al. AKT-dependent and -independent pathways mediate PTEN deletion-induced CNS axon regeneration. Cell Death Dis. 10, 203 (2019).

60

Zhang, J., Yang, D., Huang, H., Sun, Y. & Hu, Y. Coordination of necessary and permissive signals by PTEN inhibition for CNS axon regeneration. Front. Neurosci. 12, 558 (2018).

61

Miao, L. et al. mTORC1 is necessary but mTORC2 and GSK3beta are inhibitory for AKT3-induced axon regeneration in the central nervous system. Elife 5, e14908 (2016).

62

Liu, G., Detloff, M. R., Miller, K. N., Santi, L. & Houle, J. D. Exercise modulates microRNAs that affect the PTEN/mTOR pathway in rats after spinal cord injury. Exp. Neurol. 233, 447–456 (2012).

63

Yang, P., Wen, H., Ou, S., Cui, J. & Fan, D. IL-6 promotes regeneration and functional recovery after cortical spinal tract injury by reactivating intrinsic growth program of neurons and enhancing synapse formation. Exp. Neurol. 236, 19–27 (2012).

64

Zareen, N. et al. Stimulation-dependent remodeling of the corticospinal tract requires reactivation of growth-promoting developmental signaling pathways. Exp. Neurol. 307, 133–144 (2018).

65

Blanquie, O. & Bradke, F. Cytoskeleton dynamics in axon regeneration. Curr. Opin. Neurobiol. 51, 60–69 (2018).

66

Tom, V. J., Steinmetz, M. P., Miller, J. H., Doller, C. M. & Silver, J. Studies on the development and behavior of the dystrophic growth cone, the hallmark of regeneration failure, in an in vitro model of the glial scar and after spinal cord injury. J. Neurosci. 24, 6531–6539 (2004).

67

Ertürk, A., Hellal, F., Enes, J. & Bradke, F. Disorganized microtubules underlie the formation of retraction bulbs and the failure of axonal regeneration. J. Neurosci. 27, 9169–9180 (2007).

68

Conde, C. & Cáceres, A. Microtubule assembly, organization and dynamics in axons and dendrites. Nat. Rev. Neurosci. 10, 319–332 (2009).

69

Jiang, H., Guo, W., Liang, X. & Rao, Y. Both the establishment and the maintenance of neuronal polarity require active mechanisms: critical roles of GSK-3beta and its upstream regulators. Cell 120, 123–135 (2005).

70

Zhou, F. Q., Zhou, J., Dedhar, S., Wu, Y. H. & Snider, W. D. NGF-induced axon growth is mediated by localized inactivation of GSK-3beta and functions of the microtubule plus end binding protein APC. Neuron 42, 897–912 (2004).

71

Schaefer, A. W. et al. Coordination of actin filament and microtubule dynamics during neurite outgrowth. Dev. Cell 15, 146–162 (2008).

72

Zhang, X. F., Schaefer, A. W., Burnette, D. T., Schoonderwoert, V. T. & Forscher, P. Rho-dependent contractile responses in the neuronal growth cone are independent of classical peripheral retrograde actin flow. Neuron 40, 931–944 (2003).

73

He, Z. & Jin, Y. Intrinsic control of axon regeneration. Neuron 90, 437–451 (2016).

74

Han, Q. et al. Restoring cellular energetics promotes axonal regeneration and functional recovery after spinal cord injury. Cell Metab. 31, 623–641.e628 (2020).

75

McEwen, M. L., Sullivan, P. G., Rabchevsky, A. G. & Springer, J. E. Targeting mitochondrial function for the treatment of acute spinal cord injury. Neurotherapeutics 8, 168–179 (2011).

76

Jia, Z. Q. et al. Time representation of mitochondrial morphology and function after acute spinal cord injury. Neural Regen. Res. 11, 137–143 (2016).

77

Han, S. M., Baig, H. S. & Hammarlund, M. Mitochondria localize to injured axons to support regeneration. Neuron 92, 1308–1323 (2016).

78

Zhou, B. et al. Facilitation of axon regeneration by enhancing mitochondrial transport and rescuing energy deficits. J. Cell Biol. 214, 103–119 (2016).

79

Wang, C. et al. An injectable heparin-Laponite hydrogel bridge FGF4 for spinal cord injury by stabilizing microtubule and improving mitochondrial function. Theranostics 9, 7016–7032 (2019).

80

Wang, X. et al. PINK1 and Parkin target Miro for phosphorylation and degradation to arrest mitochondrial motility. Cell 147, 893–906 (2011).

81

Yang, W. & Hekimi, S. A mitochondrial superoxide signal triggers increased longevity in Caenorhabditis elegans. PLoS Biol. 8, e1000556 (2010).

82

Yee, C., Yang, W. & Hekimi, S. The intrinsic apoptosis pathway mediates the pro-longevity response to mitochondrial ROS in C. elegans. Cell 157, 897–909 (2014).

83

Sheng, Z. H. The interplay of axonal energy homeostasis and mitochondrial trafficking and anchoring. Trends Cell Biol. 27, 403–416 (2017).

84

Chung, W. S. et al. Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways. Nature 504, 394–400 (2013).

85

Allen, N. J. et al. Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature 486, 410–414 (2012).

86

Kucukdereli, H. et al. Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc. Natl. Acad. Sci. USA 108, E440–E449 (2011).

87

Allen, N. J. & Barres, B. A. Neuroscience: Glia - more than just brain glue. Nature 457, 675–677 (2009).

88

Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

89

Bush, T. G. et al. Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron 23, 297–308 (1999).

90

Wilhelmsson, U. et al. Redefining the concept of reactive astrocytes as cells that remain within their unique domains upon reaction to injury. Proc. Natl. Acad. Sci. USA 103, 17513–17518 (2006).

91

Hara, M. et al. Interaction of reactive astrocytes with type Ⅰ collagen induces astrocytic scar formation through the integrin-N-cadherin pathway after spinal cord injury. Nat. Med. 23, 818–828 (2017).

92

Silver, J. & Miller, J. H. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 5, 146–156 (2004).

93

Mikami, T. & Kitagawa, H. Biosynthesis and function of chondroitin sulfate. Biochim. Biophys. Acta 1830, 4719–4733 (2013).

94

Fawcett, J. W. The extracellular matrix in plasticity and regeneration after CNS injury and neurodegenerative disease. Prog. Brain Res. 218, 213–226 (2015).

95

Bradbury, E. J. et al. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 416, 636–640 (2002).

96

Wang, D., Ichiyama, R. M., Zhao, R., Andrews, M. R. & Fawcett, J. W. Chondroitinase combined with rehabilitation promotes recovery of forelimb function in rats with chronic spinal cord injury. J. Neurosci. 31, 9332–9344 (2011).

97

Brambilla, R. et al. Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J. Exp. Med. 202, 145–156 (2005).

98

Faulkner, J. R. et al. Reactive astrocytes protect tissue and preserve function after spinal cord injury. J. Neurosci. 24, 2143–2155 (2004).

99

Wanner, I. B. et al. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J. Neurosci. 33, 12870–12886 (2013).

100

Anderson, M. A. et al. Astrocyte scar formation aids central nervous system axon regeneration. Nature 532, 195–200 (2016).

101

Nathan, F. M. & Li, S. Environmental cues determine the fate of astrocytes after spinal cord injury. Neural Regen. Res. 12, 1964–1970 (2017).

102

Armulik, A., Genove, G. & Betsholtz, C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 21, 193–215 (2011).

103

Hall, C. N. et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508, 55–60 (2014).

104

Bell, R. D. et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 68, 409–427 (2010).

105

Silver, J. The glial scar is more than just astrocytes. Exp. Neurol. 286, 147–149 (2016).

106

Dias, D. O. et al. Reducing pericyte-derived scarring promotes recovery after spinal cord injury. Cell 173, 153–165.e122 (2018).

107

Gurtner, G. C., Werner, S., Barrandon, Y. & Longaker, M. T. Wound repair and regeneration. Nature 453, 314–321 (2008).

108

Zhu, Y., Soderblom, C., Trojanowsky, M., Lee, D. H. & Lee, J. K. Fibronectin Matrix Assembly after Spinal Cord Injury. J. Neurotrauma 32, 1158–1167 (2015).

109

Goritz, C. et al. A pericyte origin of spinal cord scar tissue. Science 333, 238–242 (2011).

110

Hesp, Z. C. et al. Proliferating NG2-cell-dependent angiogenesis and scar formation alter axon growth and functional recovery after spinal cord injury in mice. J. Neurosci. 38, 1366–1382 (2018).

111

Sauer, R. S. et al. Blood-spinal cord barrier breakdown and pericyte deficiency in peripheral neuropathy. Ann. N. Y. Acad. Sci. 1405, 71–88 (2017).

112

Stark, K. et al. Capillary and arteriolar pericytes attract innate leukocytes exiting through venules and ‘instruct’ them with pattern-recognition and motility programs. Nat. Immunol. 14, 41–51 (2013).

113

Karow, M. et al. Reprogramming of pericyte-derived cells of the adult human brain into induced neuronal cells. Cell Stem Cell 11, 471–476 (2012).

114

De La Fuente, A. G. et al. Pericytes stimulate oligodendrocyte progenitor cell differentiation during CNS remyelination. Cell Rep. 20, 1755–1764 (2017).

115

Orr, M. B. & Gensel, J. C. Spinal cord injury scarring and inflammation: therapies targeting glial and inflammatory responses. Neurotherapeutics 15, 541–553 (2018).

116

David, S. & Kroner, A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat. Rev. Neurosci. 12, 388–399 (2011).

117

Hines, D. J., Hines, R. M., Mulligan, S. J. & Macvicar, B. A. Microglia processes block the spread of damage in the brain and require functional chloride channels. Glia 57, 1610–1618 (2009).

118

Perrin, F. E., Lacroix, S., Aviles-Trigueros, M. & David, S. Involvement of monocyte chemoattractant protein-1, macrophage inflammatory protein-1alpha and interleukin-1beta in Wallerian degeneration. Brain 128, 854–866 (2005).

119

Haynes, S. E. et al. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 9, 1512–1519 (2006).

120

Martinez, F. O., Sica, A., Mantovani, A. & Locati, M. Macrophage activation and polarization. Front. Biosci. 13, 453–461 (2008).

121

Murray, P. J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

122

Ginhoux, F. & Prinz, M. Origin of microglia: current concepts and past controversies. Cold Spring Harb. Perspect. Biol. 7, a020537 (2015).

123

Greenhalgh, A. D. & David, S. Differences in the phagocytic response of microglia and peripheral macrophages after spinal cord injury and its effects on cell death. J. Neurosci. 34, 6316–6322 (2014).

124

Beck, K. D. et al. Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain 133, 433–447 (2010).

125

Conti, A. et al. Nitric oxide in the injured spinal cord: synthases cross-talk, oxidative stress and inflammation. Brain Res. Rev. 54, 205–218 (2007).

126

Bains, M. & Hall, E. D. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim. Biophys. Acta 1822, 675–684 (2012).

127

Lawrence, T. & Natoli, G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat. Rev. Immunol. 11, 750–761 (2011).

128

Yang, L. et al. Early expression and cellular localization of proinflammatory cytokines interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha in human traumatic spinal cord injury. Spine (Philos. Pa 1976) 29, 966–971 (2004).

129

Ferguson, A. R. et al. Cell death after spinal cord injury is exacerbated by rapid TNF alpha-induced trafficking of GluR2-lacking AMPARs to the plasma membrane. J. Neurosci. 28, 11391–11400 (2008).

130

Takahashi, K., Rochford, C. D. & Neumann, H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J. Exp. Med. 201, 647–657 (2005).

131

Shechter, R. et al. Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice. PLoS Med. 6, e1000113 (2009).

132

Crowe, M. J., Bresnahan, J. C., Shuman, S. L., Masters, J. N. & Crowe, M. S. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat. Med. 3, 73–76 (1997).

133

Crowe, M. J., Bresnahan, J. C., Shuman, S. L., Masters, J. N. & Beattie, M. S. Apoptosis and delayed degeneration after spinal cord injury in rats and monkeys. Nat. Med. 3, 73–76 (1997).

134

Li, G. L., Farooque, M., Holtz, A. & Olsson, Y. Apoptosis of oligodendrocytes occurs for long distances away from the primary injury after compression trauma to rat spinal cord. Acta Neuropathol. 98, 473–480 (1999).

135

Fan, H. et al. Involvement of endoplasmic reticulum stress in the necroptosis of microglia/macrophages after spinal cord injury. Neuroscience 311, 362–373 (2015).

136

Fan, H. et al. Quercetin prevents necroptosis of oligodendrocytes by inhibiting macrophages/microglia polarization to M1 phenotype after spinal cord injury in rats. J. Neuroinflammation 16, 206 (2019).

137

Garcia, E., Aguilar-Cevallos, J., Silva-Garcia, R. & Ibarra, A. Cytokine and growth factor activation in vivo and in vitro after spinal cord injury. Mediators Inflamm. 2016, 9476020 (2016).

138

Salter, M. G. & Fern, R. NMDA receptors are expressed in developing oligodendrocyte processes and mediate injury. Nature 438, 1167–1171 (2005).

139

Oka, A., Belliveau, M. J., Rosenberg, P. A. & Volpe, J. J. Vulnerability of oligodendroglia to glutamate: pharmacology, mechanisms, and prevention. J. Neurosci. 13, 1441–1453 (1993).

140

Liu, D., Liu, J., Sun, D., Alcock, N. W. & Wen, J. Spinal cord injury increases iron levels: catalytic production of hydroxyl radicals. Free Radic. Biol. Med. 34, 64–71 (2003).

141

Gao, M. et al. Ferroptosis is an autophagic cell death process. Cell Res. 26, 1021–1032 (2016).

142

Zhang, X. et al. Cytokine toxicity to oligodendrocyte precursors is mediated by iron. Glia 52, 199–208 (2005).

143

Fan, B. Y. et al. Liproxstatin-1 is an effective inhibitor of oligodendrocyte ferroptosis induced by inhibition of glutathione peroxidase 4. Neural Regen. Res. 16, 561–566 (2021).

144

Chen, K. B. et al. Tumor necrosis factor-α antagonist reduces apoptosis of neurons and oligodendroglia in rat spinal cord injury. Spine (Philos. Pa 1976) 36, 1350–1358 (2011).

145

Lee, Y. B. et al. Role of tumor necrosis factor-alpha in neuronal and glial apoptosis after spinal cord injury. Exp. Neurol. 166, 190–195 (2000).

146

Yan, P. et al. Expression of the type 1 and type 2 receptors for tumor necrosis factor after traumatic spinal cord injury in adult rats. Exp. Neurol. 183, 286–297 (2003).

147

Yan, P. et al. Cellular localization of tumor necrosis factor-alpha following acute spinal cord injury in adult rats. J. Neurotrauma 18, 563–568 (2001).

148

Fan, B. et al. Microenvironment Imbalance of spinal cord injury. Cell Transplant. 27, 853–866 (2018).

149

Kanno, H., Ozawa, H., Sekiguchi, A., Yamaya, S. & Itoi, E. Induction of autophagy and autophagic cell death in damaged neural tissue after acute spinal cord injury in mice. Spine (Philos. Pa 1976) 36, E1427–E1434 (2011).

150

Muñoz-Galdeano, T. et al. Cell specific changes of autophagy in a mouse model of contusive spinal cord injury. Front. Cell. Neurosci. 12, 164 (2018).

151

Smith, C. M., Mayer, J. A. & Duncan, I. D. Autophagy promotes oligodendrocyte survival and function following dysmyelination in a long-lived myelin mutant. J. Neurosci. 33, 8088–8100 (2013).

152

Bankston, A. N. et al. Autophagy is essential for oligodendrocyte differentiation, survival, and proper myelination. Glia 67, 1745–1759 (2019).

153

Saraswat Ohri, S. et al. Blocking autophagy in oligodendrocytes limits functional recovery after spinal cord injury. J. Neurosci. 38, 5900–5912 (2018).

154

GrandPré, T., Nakamura, F., Vartanian, T. & Strittmatter, S. M. Identification of the Nogo inhibitor of axon regeneration as a Reticulon protein. Nature 403, 439–444 (2000).

155

Huber, A. B. & Schwab, M. E. Nogo-A, a potent inhibitor of neurite outgrowth and regeneration. Biol. Chem. 381, 407–419 (2000).

156

Chen, M. S. et al. Nogo-A is a myelin-associated neurite outgrowth inhibitor and an antigen for monoclonal antibody IN-1. Nature 403, 434–439 (2000).

157

Schwab, M. Functions of Nogo proteins and their receptors in the nervous system. Nat. Rev. Neurosci. 11, 799–811 (2010).

158

Liu, B., Fournier, A., GrandPré, T. & Strittmatter, S. Myelin-associated glycoprotein as a functional ligand for the Nogo-66 receptor. Science 297, 1190–1193 (2002).

159

Domeniconi, M. et al. Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth. Neuron 35, 283–290 (2002).

160

Wang, K. C., Kim, J. A., Sivasankaran, R., Segal, R. & He, Z. P75 interacts with the Nogo receptor as a co-receptor for Nogo, MAG and OMgp. Nature 420, 74–78 (2002).

161

McTigue, D. M., Popovich, P. G., Morgan, T. E. & Stokes, B. T. Localization of transforming growth factor-beta1 and receptor mRNA after experimental spinal cord injury. Exp. Neurol. 163, 220–230 (2000).

162

Tripathi, R. & McTigue, D. M. Prominent oligodendrocyte genesis along the border of spinal contusion lesions. Glia 55, 698–711 (2007).

163

Asher, R. A. et al. Two separate metalloproteinase activities are responsible for the shedding and processing of the NG2 proteoglycan in vitro. Mol. Cell. Neurosci. 29, 82–96 (2005).

164

Dou, C. L. & Levine, J. M. Inhibition of neurite growth by the NG2 chondroitin sulfate proteoglycan. J. Neurosci. 14, 7616–7628 (1994).

165

Harlow, D. E. & Macklin, W. B. Inhibitors of myelination: ECM changes, CSPGs and PTPs. Exp. Neurol. 251, 39–46 (2014).

166

McDonough, A., Hoang, A., Monterrubio, A., Greenhalgh, S. & Martínez-Cerdeño, V. Compression injury in the mouse spinal cord elicits a specific proliferative response and distinct cell fate acquisition along rostro-caudal and dorso-ventral axes. Neuroscience 254, 1–17 (2013).

167

Rabchevsky, A. G., Sullivan, P. G. & Scheff, S. W. Temporal-spatial dynamics in oligodendrocyte and glial progenitor cell numbers throughout ventrolateral white matter following contusion spinal cord injury. Glia 55, 831–843 (2007).

168

Hassannejad, Z. et al. Oligodendrogliogenesis and axon remyelination after traumatic spinal cord injuries in animal studies: a systematic review. Neuroscience 402, 37–50 (2019).

169

Assinck, P. et al. Myelinogenic plasticity of oligodendrocyte precursor cells following spinal cord contusion injury. J. Neurosci. 37, 8635–8654 (2017).

170

McTigue, D. M., Wei, P. & Stokes, B. T. Proliferation of NG2-positive cells and altered oligodendrocyte numbers in the contused rat spinal cord. J. Neurosci. 21, 3392–3400 (2001).

171

Duncan, G. J. et al. Locomotor recovery following contusive spinal cord injury does not require oligodendrocyte remyelination. Nat. Commun. 9, 3066 (2018).

172

Duncan, I. D. et al. The adult oligodendrocyte can participate in remyelination. Proc. Natl. Acad. Sci. USA 115, E11807–E11816 (2018).

173

Bunge, M. B., Bunge, R. P. & Ris, H. Ultrastructural study of remyelination in an experimental lesion in adult cat spinal cord. J. Biophys. Biochem. Cytol. 10, 67–94 (1961).

174

Guest, J. D., Hiester, E. D. & Bunge, R. P. Demyelination and Schwann cell responses adjacent to injury epicenter cavities following chronic human spinal cord injury. Exp. Neurol. 192, 384–393 (2005).

175

James, N. D. et al. Conduction failure following spinal cord injury: functional and anatomical changes from acute to chronic stages. J. Neurosci. 31, 18543–18555 (2011).

176

Nagoshi, N. et al. Schwann cell plasticity after spinal cord injury shown by neural crest lineage tracing. Glia 59, 771–784 (2011).

177

Franklin, R. J. & Blakemore, W. F. Requirements for Schwann cell migration within CNS environments: a viewpoint. Int J. Dev. Neurosci. 11, 641–649 (1993).

178

Gage, F. H. & Temple, S. Neural stem cells: generating and regenerating the brain. Neuron 80, 588–601 (2013).

179

Johansson, C. B. et al. Identification of a neural stem cell in the adult mammalian central nervous system. Cell 96, 25–34 (1999).

180

Liu, S. & Chen, Z. Employing endogenous NSCs to promote recovery of spinal cord injury. Stem Cells Int. 2019, 1958631 (2019).

181

Barnabé-Heider, F. et al. Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell 7, 470–482 (2010).

182

Liu, Y. et al. Endogenous neural stem cells in central canal of adult rats acquired limited ability to differentiate into neurons following mild spinal cord injury. Int J. Clin. Exp. Pathol. 8, 3835–3842 (2015).

183

Meletis, K. et al. Spinal cord injury reveals multilineage differentiation of ependymal cells. PLoS Biol. 6, e182 (2008).

184

Llorens-Bobadilla, E. et al. A latent lineage potential in resident neural stem cells enables spinal cord repair. Science 370, eabb8795 (2020).

185

Zholudeva, L. V. et al. The neuroplastic and therapeutic potential of spinal interneurons in the injured spinal cord. Trends Neurosci. 41, 625–639 (2018).

186

Lu, D. C., Niu, T. & Alaynick, W. A. Molecular and cellular development of spinal cord locomotor circuitry. Front. Mol. Neurosci. 8, 25 (2015).

187

Laliberte, A. M., Goltash, S., Lalonde, N. R. & Bui, T. V. Propriospinal neurons: essential elements of locomotor control in the intact and possibly the injured spinal cord. Front. Cell. Neurosci. 13, 512 (2019).

188

Zholudeva, L. V., Karliner, J. S., Dougherty, K. J. & Lane, M. A. Anatomical recruitment of spinal V2a interneurons into phrenic motor circuitry after high cervical spinal cord injury. J. Neurotrauma 34, 3058–3065 (2017).

189

Rank, M. M., Flynn, J. R., Galea, M. P., Callister, R. & Callister, R. J. Electrophysiological characterization of spontaneous recovery in deep dorsal horn interneurons after incomplete spinal cord injury. Exp. Neurol. 271, 468–478 (2015).

190

Bareyre, F. M. et al. The injured spinal cord spontaneously forms a new intraspinal circuit in adult rats. Nat. Neurosci. 7, 269–277 (2004).

191

Filli, L. et al. Bridging the gap: a reticulo-propriospinal detour bypassing an incomplete spinal cord injury. J. Neurosci. 34, 13399–13410 (2014).

192

May, Z. et al. Following spinal cord injury transected reticulospinal tract axons develop new collateral inputs to spinal interneurons in parallel with locomotor recovery. Neural Plast. 2017, 1932875 (2017).

193

Liu, Z.-H. et al. A single bolus of docosahexaenoic acid promotes neuroplastic changes in the innervation of spinal cord interneurons and motor neurons and improves functional recovery after spinal cord injury. J. Neurosci. 35, 12733–12752 (2015).

194

Dru, A. B. & Hoh, D. J. Activating spinal interneurons for neural repair after spinal cord injury. World Neurosurg. 84, 1185–1188 (2015).

195

Courtine, G. et al. Recovery of supraspinal control of stepping via indirect propriospinal relay connections after spinal cord injury. Nat. Med. 14, 69–74 (2008).

196

Takeoka, A., Vollenweider, I., Courtine, G. & Arber, S. Muscle spindle feedback directs locomotor recovery and circuit reorganization after spinal cord injury. Cell 159, 1626–1639 (2014).

197

Bui, T. V., Stifani, N., Akay, T. & Brownstone, R. M. Spinal microcircuits comprising dI3 interneurons are necessary for motor functional recovery following spinal cord transection. Elife 5, e21715 (2016).

198

Ganzer, P. D. et al. Restoring the sense of touch using a sensorimotor demultiplexing neural interface. Cell 181, 763–773.e12 (2020).

199

Hayashi, M. et al. Graded arrays of spinal and supraspinal V2a interneuron subtypes underlie forelimb and hindlimb motor control. Neuron 97, 869–884.e865 (2018).

200

Dougherty, K. et al. Locomotor rhythm generation linked to the output of spinal shox2 excitatory interneurons. Neuron 80, 920–933 (2013).

201

Caldeira, V., Dougherty, K., Borgius, L. & Kiehn, O. Spinal Hb9::Cre-derived excitatory interneurons contribute to rhythm generation in the mouse. Sci. Rep. 7, 41369 (2017).

202

Romer, S. H. et al. Accessory respiratory muscles enhance ventilation in ALS model mice and are activated by excitatory V2a neurons. Exp. Neurol. 287, 192–204 (2017).

203

Gwak, Y. S. & Hulsebosch, C. E. GABA and central neuropathic pain following spinal cord injury. Neuropharmacology 60, 799–808 (2011).

204

Grau, J. et al. Metaplasticity and behavior: how training and inflammation affect plastic potential within the spinal cord and recovery after injury. Front. Neural Circuits 8, 100 (2014).

205

Ramírez-Jarquín, U. & Tapia, R. Excitatory and inhibitory neuronal circuits in the spinal cord and their role in the control of motor neuron function and degeneration. ACS Chem. Neurosci. 9, 211–216 (2018).

206

Huang, H. et al. Clinical neurorestorative therapeutic guidelines for spinal cord injury (IANR/CANR version 2019). J. Orthop. Translat. 20, 14–24 (2020).

207

Shultz, R. B. & Zhong, Y. Minocycline targets multiple secondary injury mechanisms in traumatic spinal cord injury. Neural Regen. Res. 12, 702–713 (2017).

208

Lee, S. M. et al. Minocycline reduces cell death and improves functional recovery after traumatic spinal cord injury in the rat. J. Neurotrauma 20, 1017–1027 (2003).

209

Casha, S. et al. Results of a phase Ⅱ placebo-controlled randomized trial of minocycline in acute spinal cord injury. Brain 135, 1224–1236 (2012).

210

Nagoshi, N., Nakashima, H. & Fehlings, M. G. Riluzole as a neuroprotective drug for spinal cord injury: from bench to bedside. Molecules 20, 7775–7789 (2015).

211

Grossman, R. G. et al. A prospective, multicenter, phase Ⅰ matched-comparison group trial of safety, pharmacokinetics, and preliminary efficacy of riluzole in patients with traumatic spinal cord injury. J. Neurotrauma 31, 239–255 (2014).

212

Tetreault, L. A., Zhu, M. P., Wilson, J. R., Karadimas, S. K. & Fehlings, M. G. The impact of riluzole on neurobehavioral outcomes in preclinical models of traumatic and nontraumatic spinal cord injury: results from a systematic review of the literature. Glob. Spine J. 10, 216–229 (2020).

213

Khorasanizadeh, M., Eskian, M., Vaccaro, A. R. & Rahimi-Movaghar, V. Granulocyte colony-stimulating factor (G-CSF) for the treatment of spinal cord injury. CNS Drugs 31, 911–937 (2017).

214

Inada, T. et al. Multicenter prospective nonrandomized controlled clinical trial to prove neurotherapeutic effects of granulocyte colony-stimulating factor for acute spinal cord injury: analyses of follow-up cases after at least 1 year. Spine (Philos. Pa 1976) 39, 213–219 (2014).

215

Azizi, M. et al. ChABC-loaded PLGA nanoparticles: a comprehensive study on biocompatibility, functional recovery, and axonal regeneration in animal model of spinal cord injury. Int J. Pharm. 577, 119037 (2020).

216

Pakulska, M. M., Tator, C. H. & Shoichet, M. S. Local delivery of chondroitinase ABC with or without stromal cell-derived factor 1α promotes functional repair in the injured rat spinal cord. Biomaterials 134, 13–21 (2017).

217

Wu, D. et al. Combining constitutively active rheb expression and chondroitinase promotes functional axonal regeneration after cervical spinal cord injury. Mol. Ther. 25, 2715–2726 (2017).

218

Tator, C. H. et al. Translational potential of preclinical trials of neuroprotection through pharmacotherapy for spinal cord injury. J. Neurosurg. Spine 17, 157–229 (2012).

219

Thibault-Halman, G. et al. Predicting recruitment feasibility for acute spinal cord injury clinical trials in Canada using national registry data. J. Neurotrauma 34, 599–606 (2017).

220

Ulndreaj, A., Badner, A. & Fehlings, M. G. Promising neuroprotective strategies for traumatic spinal cord injury with a focus on the differential effects among anatomical levels of injury. F1000Res 6, 1907 (2017).

221

Fehlings, M. G. et al. A phase Ⅰ/Ⅱa clinical trial of a recombinant Rho protein antagonist in acute spinal cord injury. J. Neurotrauma 28, 787–796 (2011).

222

Freund, P. et al. Anti-Nogo-A antibody treatment enhances sprouting of corticospinal axons rostral to a unilateral cervical spinal cord lesion in adult macaque monkey. J. Comp. Neurol. 502, 644–659 (2007).

223

Kucher, K. et al. First-in-man intrathecal application of neurite growth-promoting anti-nogo-A Antibodies in Acute Spinal Cord Injury. Neurorehabil Neural Repair 32, 578–589 (2018).

224

Wu, J. C., Huang, W. C., Tsai, Y. A., Chen, Y. C. & Cheng, H. Nerve repair using acidic fibroblast growth factor in human cervical spinal cord injury: a preliminary Phase Ⅰ clinical study. J. Neurosurg. Spine 8, 208–214 (2008).

225

Zhou, Y., Wang, Z., Li, J., Li, X. & Xiao, J. Fibroblast growth factors in the management of spinal cord injury. J. Cell. Mol. Med. 22, 25–37 (2018).

226

Hall, E. D. & Braughler, J. M. Glucocorticoid mechanisms in acute spinal cord injury: a review and therapeutic rationale. Surg. Neurol. 18, 320–327 (1982).

227

Hall, E. D. & Braughler, J. M. Effects of intravenous methylprednisolone on spinal cord lipid peroxidation and Na+ + K+)-ATPase activity. Dose-response analysis during 1st hour after contusion injury in the cat. J. Neurosurg 57, 247–253 (1982).

228

Bracken, M. B. et al. Efficacy of methylprednisolone in acute spinal cord injury. JAMA 251, 45–52 (1984).

229

Bracken, M. B. et al. A randomized, controlled trial of methylprednisolone or naloxone in the treatment of acute spinal-cord injury. Results of the Second National Acute Spinal Cord Injury Study. N. Engl. J. Med. 322, 1405–1411 (1990).

230

Coleman, W. P. et al. A critical appraisal of the reporting of the National Acute Spinal Cord Injury Studies (Ⅱ and Ⅲ) of methylprednisolone in acute spinal cord injury. J. Spinal Disord. 13, 185–199 (2000).

231

Hurlbert, R. J. Methylprednisolone for acute spinal cord injury: an inappropriate standard of care. J. Neurosurg. 93, 1–7 (2000).

232

Bracken, M. B. et al. Administration of methylprednisolone for 24 or 48 h or tirilazad mesylate for 48 h in the treatment of acute spinal cord injury. Results of the Third National Acute Spinal Cord Injury Randomized Controlled Trial. National Acute Spinal Cord Injury Study. JAMA 277, 1597–1604 (1997).

233

Bracken, M. B. Steroids for acute spinal cord injury. Cochrane Database Syst. Rev. 1, Cd001046 (2012).

234

Fehlings, M. G. et al. A clinical practice guideline for the management of patients with acute spinal cord injury: recommendations on the use of methylprednisolone sodium succinate. Glob. Spine J. 7, 203s–211s (2017).

235

Evaniew, N. et al. Methylprednisolone for the treatment of patients with acute spinal cord injuries: a propensity score-matched cohort study from a Canadian Multi-Center Spinal Cord Injury Registry. J. Neurotrauma 32, 1674–1683 (2015).

236

Liu, L. J. W., Rosner, J. & Cragg, J. J. Journal Club: high-dose methylprednisolone for acute traumatic spinal cord injury: a meta-analysis. Neurology 95, 272–274 (2020).

237

Roquilly, A. et al. French recommendations for the management of patients with spinal cord injury or at risk of spinal cord injury. Anaesth. Crit. Care Pain. Med. 39, 279–289 (2020).

238

Walters, B. C. et al. Guidelines for the management of acute cervical spine and spinal cord injuries: 2013 update. Neurosurgery 60, 82–91 (2013).

239

Wells, J. E., Hurlbert, R. J., Fehlings, M. G. & Yong, V. W. Neuroprotection by minocycline facilitates significant recovery from spinal cord injury in mice. Brain 126, 1628–1637 (2003).

240

Festoff, B. W. et al. Minocycline neuroprotects, reduces microgliosis, and inhibits caspase protease expression early after spinal cord injury. J. Neurochem. 97, 1314–1326 (2006).

241

Teng, Y. D. et al. Minocycline inhibits contusion-triggered mitochondrial cytochrome c release and mitigates functional deficits after spinal cord injury. Proc. Natl. Acad. Sci. USA 101, 3071–3076 (2004).

242

Pinzon, A. et al. A re-assessment of minocycline as a neuroprotective agent in a rat spinal cord contusion model. Brain Res. 1243, 146–151 (2008).

243

Lee, J. H. et al. Lack of neuroprotective effects of simvastatin and minocycline in a model of cervical spinal cord injury. Exp. Neurol. 225, 219–230 (2010).

244

Stutzmann, J. M., Pratt, J., Boraud, T. & Gross, C. The effect of riluzole on post-traumatic spinal cord injury in the rat. Neuroreport 7, 387–392 (1996).

245

Kitzman, P. H. Effectiveness of riluzole in suppressing spasticity in the spinal cord injured rat. Neurosci. Lett. 455, 150–153 (2009).

246

Yoon, S. H. 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 25, 2066–2073 (2007).

247

Takahashi, H. et al. Neuroprotective therapy using granulocyte colony-stimulating factor for acute spinal cord injury: a phase Ⅰ/Ⅱa clinical trial. Eur. Spine J. 21, 2580–2587 (2012).

248

Di Gregorio, F., Ferrari, G., Marini, P., Siliprandi, R. & Gorio, A. The influence of gangliosides on neurite growth and regeneration. Neuropediatrics 15, 93–96 (1984).

249

Bose, B., Osterholm, J. L. & Kalia, M. Ganglioside-induced regeneration and reestablishment of axonal continuity in spinal cord-transected rats. Neurosci. Lett. 63, 165–169 (1986).

250

Geisler, F. H., Dorsey, F. C. & Coleman, W. P. Recovery of motor function after spinal-cord injury−a randomized, placebo-controlled trial with GM-1 ganglioside. N. Engl. J. Med. 324, 1829–1838 (1991).

251

Geisler, F. H., Coleman, W. P., Grieco, G. & Poonian, D. The Sygen multicenter acute spinal cord injury study. Spine (Philos. Pa 1976) 26, S87–S98 (2001).

252

Jevans, B. et al. Combined treatment with enteric neural stem cells and chondroitinase ABC reduces spinal cord lesion pathology. Stem Cell Res. Ther. 12, 10 (2021).

253

Fouad, K. et al. Combining Schwann cell bridges and olfactory-ensheathing glia grafts with chondroitinase promotes locomotor recovery after complete transection of the spinal cord. J. Neurosci. 25, 1169–1178 (2005).

254

Dergham, P. et al. Rho signaling pathway targeted to promote spinal cord repair. J. Neurosci. 22, 6570–6577 (2002).

255

Stern, S. et al. RhoA drives actin compaction to restrict axon regeneration and astrocyte reactivity after CNS injury. Neuron 109, 3436–3455.e3439 (2021).

256

Schwab, M. E. & Caroni, P. Oligodendrocytes and CNS myelin are nonpermissive substrates for neurite growth and fibroblast spreading in vitro. J. Neurosci. 8, 2381–2393 (1988).

257

Schnell, L. & Schwab, M. E. Axonal regeneration in the rat spinal cord produced by an antibody against myelin-associated neurite growth inhibitors. Nature 343, 269–272 (1990).

258

Fouad, K., Klusman, I. & Schwab, M. E. Regenerating corticospinal fibers in the Marmoset (Callitrix jacchus) after spinal cord lesion and treatment with the anti-Nogo-A antibody IN-1. Eur. J. Neurosci. 20, 2479–2482 (2004).

259

Logan, A. & Berry, M. Transforming growth factor-beta 1 and basic fibroblast growth factor in the injured CNS. Trends Pharmacol. Sci. 14, 337–342 (1993).

260

Ribeiro-Resende, V. T., Carrier-Ruiz, A., Lemes, R. M., Reis, R. A. & Mendez-Otero, R. Bone marrow-derived fibroblast growth factor-2 induces glial cell proliferation in the regenerating peripheral nervous system. Mol. Neurodegener. 7, 34 (2012).

261

Hohn, A., Leibrock, J., Bailey, K. & Barde, Y. A. Identification and characterization of a novel member of the nerve growth factor/brain-derived neurotrophic factor family. Nature 344, 339–341 (1990).

262
A controlled trial of recombinant methionyl human BDNF in ALS: the BDNF Study Group (Phase Ⅲ). Neurology 52, 1427–1433 (1999).
263

Tuszynski, M. H. et al. NT-3 gene delivery elicits growth of chronically injured corticospinal axons and modestly improves functional deficits after chronic scar resection. Exp. Neurol. 181, 47–56 (2003).

264

Helgren, M. E. et al. Neurotrophin-3 administration attenuates deficits of pyridoxine-induced large-fiber sensory neuropathy. J. Neurosci. 17, 372–382 (1997).

265

Gao, W. Q. et al. Neurotrophin-3 reverses experimental cisplatin-induced peripheral sensory neuropathy. Ann. Neurol. 38, 30–37 (1995).

266

Levi-Montalcini, R. & Hamburger, V. Selective growth stimulating effects of mouse sarcoma on the sensory and sympathetic nervous system of the chick embryo. J. Exp. Zool. 116, 321–361 (1951).

267

Grill, R. J., Blesch, A. & Tuszynski, M. H. Robust growth of chronically injured spinal cord axons induced by grafts of genetically modified NGF-secreting cells. Exp. Neurol. 148, 444–452 (1997).

268

Tuszynski, M. H. et al. Nerve growth factor delivery by gene transfer induces differential outgrowth of sensory, motor, and noradrenergic neurites after adult spinal cord injury. Exp. Neurol. 137, 157–173 (1996).

269

Saadeh, Y. S. et al. The impact of blood pressure management after spinal cord injury: a systematic review of the literature. Neurosurg. Focus 43, E20 (2017).

270

Wilson, J. R., Forgione, N. & Fehlings, M. G. Emerging therapies for acute traumatic spinal cord injury. CMAJ 185, 485–492 (2013).

271

Readdy, W. J. & Dhall, S. S. Vasopressor administration in spinal cord injury: should we apply a universal standard to all injury patterns? Neural Regen. Res. 11, 420–421 (2016).

272

Hawryluk, G. et al. Mean arterial blood pressure correlates with neurological recovery after human spinal cord injury: analysis of high frequency physiologic data. J. Neurotrauma 32, 1958–1967 (2015).

273

Gunnarsson, T. & Fehlings, M. G. Acute neurosurgical management of traumatic brain injury and spinal cord injury. Curr. Opin. Neurol. 16, 717–723 (2003).

274

Karsy, M. & Hawryluk, G. Pharmacologic management of acute spinal cord injury. Neurosurg. Clin. N. Am. 28, 49–62 (2017).

275

Kanekiyo, K. et al. Effects of multiple injection of bone marrow mononuclear cells on spinal cord injury of rats. J. Neurotrauma 34, 3003–3011 (2017).

276

Ritfeld, G. J. et al. The role of brain-derived neurotrophic factor in bone marrow stromal cell-mediated spinal cord repair. Cell Transplant. 24, 2209–2220 (2015).

277

Jin, M. C., Medress, Z. A., Azad, T. D., Doulames, V. M. & Veeravagu, A. Stem cell therapies for acute spinal cord injury in humans: a review. Neurosurg. Focus 46, E10 (2019).

278

Vismara, I., Papa, S., Rossi, F., Forloni, G. & Veglianese, P. Current options for cell therapy in spinal cord injury. Trends Mol. Med. 23, 831–849 (2017).

279

Assinck, P., Duncan, G. J., Hilton, B. J., Plemel, J. R. & Tetzlaff, W. Cell transplantation therapy for spinal cord injury. Nat. Neurosci. 20, 637–647 (2017).

280

Gong, Z. et al. Stem cell transplantation: a promising therapy for spinal cord injury. Curr. Stem Cell Res. Ther. 15, 321–331 (2020).

281

Cofano, F. et al. Mesenchymal stem cells for spinal cord injury: current options, limitations, and future of cell therapy. Int. J. Mol. Sci. 20, 2698 (2019).

282

Oh, S. K. et al. A phase Ⅲ clinical trial showing limited efficacy of autologous mesenchymal stem cell therapy for spinal cord injury. Neurosurgery 78, 436–447 (2016).

283

Anderson, K. D. et al. Safety of autologous human schwann cell transplantation in subacute thoracic spinal cord injury. J. Neurotrauma 34, 2950–2963 (2017).

284

Curtis, E. et al. A first-in-human, phase Ⅰ study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell 22, 941–950 (2018).

285

Levi, A. D. et al. Clinical Outcomes from a Multi-Center Study of Human Neural Stem Cell Transplantation in Chronic Cervical Spinal Cord Injury. J. Neurotrauma 36, 891–902 (2019).

286

Zhu, H. et al. Phase Ⅰ-Ⅱ clinical trial assessing safety and efficacy of umbilical cord blood mononuclear cell transplant therapy of chronic complete spinal cord injury. Cell Transplant. 25, 1925–1943 (2016).

287

Tabakow, P. et al. Transplantation of autologous olfactory ensheathing cells in complete human spinal cord injury. Cell Transplant. 22, 1591–1612 (2013).

288

Chang, D. J. et al. Therapeutic effect of BDNF-overexpressing human neural stem cells (F3.BDNF) in a contusion model of spinal cord injury in rats. Int. J. Mol. Sci. 22, 6970 (2021).

289

Ying, Y. et al. Hypoxia response element-directed expression of aFGF in neural stem cells promotes the recovery of spinal cord injury and attenuates SCI-Induced Apoptosis. Front. Cell Dev. Biol. 9, 693694 (2021).

290

Fan, W. L. et al. Transplantation of hypoxic preconditioned neural stem cells benefits functional recovery via enhancing neurotrophic secretion after spinal cord injury in rats. J. Cell Biochem. 119, 4339–4351 (2018).

291

Zou, Y. et al. Aligned collagen scaffold combination with human spinal cord-derived neural stem cells to improve spinal cord injury repair. Biomater. Sci. 8, 5145–5156 (2020).

292

Chen, D., Hu, S., Liu, J. & Li, S. E-cadherin regulates biological behaviors of neural stem cells and promotes motor function recovery following spinal cord injury. Exp. Ther. Med. 17, 2061–2070 (2019).

293

Cusimano, M. et al. Selective killing of spinal cord neural stem cells impairs locomotor recovery in a mouse model of spinal cord injury. J. Neuroinflammation 15, 58 (2018).

294

Ogawa, Y. 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. 69, 925–933 (2002).

295

Mothe, A. J. & Tator, C. H. Review of transplantation of neural stem/progenitor cells for spinal cord injury. Int. J. Dev. Neurosci. 31, 701–713 (2013).

296

Zhu, Y., Uezono, N., Yasui, T. & Nakashima, K. Neural stem cell therapy aiming at better functional recovery after spinal cord injury. Dev. Dyn. 247, 75–84 (2018).

297

Curtis, E. et al. A first-in-human, phase Ⅰ study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell 22, 941–950.e946 (2018).

298

de Miguel Beriain, I. What is a human embryo? A new piece in the bioethics puzzle. Croat. Med J. 55, 669–671 (2014).

299

Kobayashi, Y. 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 7, e52787 (2012).

300

Ring, K. L. et al. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell 11, 100–109 (2012).

301

Dasari, V. R., Veeravalli, K. K. & Dinh, D. H. Mesenchymal stem cells in the treatment of spinal cord injuries: a review. World J. Stem Cells 6, 120–133 (2014).

302

Fuhrmann, T. et al. Axon growth-promoting properties of human bone marrow mesenchymal stromal cells. Neurosci. Lett. 474, 37–41 (2010).

303

Crigler, L., Robey, R. C., Asawachaicharn, A., Gaupp, D. & Phinney, D. G. Human mesenchymal stem cell subpopulations express a variety of neuro-regulatory molecules and promote neuronal cell survival and neuritogenesis. Exp. Neurol. 198, 54–64 (2006).

304

Hawryluk, G. W. 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. 21, 2222–2238 (2012).

305

Neirinckx, V. et al. Concise review: Spinal cord injuries: how could adult mesenchymal and neural crest stem cells take up the challenge? Stem Cells 32, 829–843 (2014).

306

Nakajima, H. et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J. Neurotrauma 29, 1614–1625 (2012).

307

Gao, S. et al. Anti-inflammatory and anti-apoptotic effect of combined treatment with methylprednisolone and amniotic membrane mesenchymal stem cells after spinal cord injury in rats. Neurochem Res. 39, 1544–1552 (2014).

308

Dasari, V. et al. Neuronal apoptosis is inhibited by cord blood stem cells after spinal cord injury. J. Neurotrauma 26, 2057–2069 (2009).

309

Bottai, D. et al. Third trimester NG2-positive amniotic fluid cells are effective in improving repair in spinal cord injury. Exp. Neurol. 254, 121–133 (2014).

310

Oh, S. K. et al. A phase Ⅲ clinical trial showing limited efficacy of autologous mesenchymal stem cell therapy for spinal cord injury. Neurosurgery 78, 436–447 (2016).

311

Vaquero, J. et al. An approach to personalized cell therapy in chronic complete paraplegia: The Puerta de Hierro phase Ⅰ/Ⅱ clinical trial. Cytotherapy 18, 1025–1036 (2016).

312

Dai, G. et al. Transplantation of autologous bone marrow mesenchymal stem cells in the treatment of complete and chronic cervical spinal cord injury. Brain Res. 1533, 73–79 (2013).

313

El-Kheir, W. A. et al. Autologous bone marrow-derived cell therapy combined with physical therapy induces functional improvement in chronic spinal cord injury patients. Cell Transplant. 23, 729–745 (2014).

314

Duncan, I., Aguayo, A., Bunge, R. & Wood, P. Transplantation of rat Schwann cells grown in tissue culture into the mouse spinal cord. J. Neurol. Sci. 49, 241–252 (1981).

315

Lankford, K. L., Imaizumi, T., Honmou, O. & Kocsis, J. D. A quantitative morphometric analysis of rat spinal cord remyelination following transplantation of allogenic Schwann cells. J. Comp. Neurol. 443, 259–274 (2002).

316

Zhang, S., Huang, F., Gates, M. & Holmberg, E. Role of endogenous Schwann cells in tissue repair after spinal cord injury. Neural Regen. Res. 8, 177–185 (2013).

317

Bunge, M. B. & Wood, P. M. Realizing the maximum potential of Schwann cells to promote recovery from spinal cord injury. Handb. Clin. Neurol. 109, 523–540 (2012).

318

Myers, S. A., Bankston, A. N., Burke, D. A., Ohri, S. S. & Whittemore, S. R. Does the preclinical evidence for functional remyelination following myelinating cell engraftment into the injured spinal cord support progression to clinical trials? Exp. Neurol. 283, 560–572 (2016).

319

Namjoo, Z. et al. Combined effects of rat Schwann cells and 17beta-estradiol in a spinal cord injury model. Metab. Brain Dis. 33, 1229–1242 (2018).

320

Zhou, X. H. et al. Transplantation of autologous activated Schwann cells in the treatment of spinal cord injury: six cases, more than five years of follow-up. Cell Transplant. 21, S39–S47 (2012).

321

Saberi, H. et al. Safety of intramedullary Schwann cell transplantation for postrehabilitation spinal cord injuries: 2-year follow-up of 33 cases. J. Neurosurg. Spine 15, 515–525 (2011).

322

Bunge, M. B. Efficacy of Schwann cell transplantation for spinal cord repair is improved with combinatorial strategies. J. Physiol. 594, 3533–3538 (2016).

323

Okano, H. et al. Steps toward safe cell therapy using induced pluripotent stem cells. Circ. Res. 112, 523–533 (2013).

324

Bellak, T. et al. Grafted human induced pluripotent stem cells improve the outcome of spinal cord injury: modulation of the lesion microenvironment. Sci. Rep. 10, 22414 (2020).

325

Khazaei, M., Ahuja, C. S. & Fehlings, M. G. Induced pluripotent stem cells for traumatic spinal cord injury. Front. Cell Dev. Biol. 4, 152 (2016).

326

Nagoshi, N., Tsuji, O., Nakamura, M. & Okano, H. Cell therapy for spinal cord injury using induced pluripotent stem cells. Regen. Ther. 11, 75–80 (2019).

327

Kawabata, S. et al. Grafted human iPS cell-derived oligodendrocyte precursor cells contribute to robust remyelination of demyelinated axons after spinal cord injury. Stem Cell Rep. 6, 1–8 (2016).

328

Gilmour, A. D., Reshamwala, R., Wright, A. A., Ekberg, J. A. K. & St John, J. A. Optimizing olfactory ensheathing cell transplantation for spinal cord injury repair. J. Neurotrauma 37, 817–829 (2020).

329

Li, Y., Field, P. & Raisman, G. Repair of adult rat corticospinal tract by transplants of olfactory ensheathing cells. Science 277, 2000–2002 (1997).

330

Kato, T., Honmou, O., Uede, T., Hashi, K. & Kocsis, J. Transplantation of human olfactory ensheathing cells elicits remyelination of demyelinated rat spinal cord. Glia 30, 209–218 (2000).

331

Pastrana, E. et al. BDNF production by olfactory ensheathing cells contributes to axonal regeneration of cultured adult CNS neurons. Neurochem. Int. 50, 491–498 (2007).

332

Lu, J. & Ashwell, K. Olfactory ensheathing cells: their potential use for repairing the injured spinal cord. Spine 27, 887–892 (2002).

333

López-Vales, R., García-Alías, G., Forés, J., Navarro, X. & Verdú, E. Increased expression of cyclo-oxygenase 2 and vascular endothelial growth factor in lesioned spinal cord by transplanted olfactory ensheathing cells. J. Neurotrauma 21, 1031–1043 (2004).

334

Chen, L. et al. A prospective randomized double-blind clinical trial using a combination of olfactory ensheathing cells and Schwann cells for the treatment of chronic complete spinal cord injuries. Cell Transplant. 23, S35–S44 (2014).

335

Lima, C. et al. Olfactory mucosal autografts and rehabilitation for chronic traumatic spinal cord injury. Neurorehabil Neural Repair 24, 10–22 (2010).

336

Dobkin, B. H., Curt, A. & Guest, J. Cellular transplants in China: observational study from the largest human experiment in chronic spinal cord injury. Neurorehabil Neural Repair 20, 5–13 (2006).

337

Mackay-Sim, A. et al. Autologous olfactory ensheathing cell transplantation in human paraplegia: a 3-year clinical trial. Brain 131, 2376–2386 (2008).

338

Guest, J. & Dietrich, W. Commentary regarding the recent publication by Tabakow et al., “functional regeneration of supraspinal connections in a patient with transected spinal cord following transplantation of bulbar olfactory ensheathing cells with peripheral nerve bridging”. J. Neurotrauma 32, 1176–1178 (2015).

339

Erceg, S. et al. Differentiation of human embryonic stem cells to regional specific neural precursors in chemically defined medium conditions. PLoS One 3, e2122 (2008).

340

McDonald, J. et al. Transplanted embryonic stem cells survive, differentiate and promote recovery in injured rat spinal cord. Nat. Med. 5, 1410–1412 (1999).

341

Shroff, G. Human embryonic stem cell therapy in chronic spinal cord injury: a retrospective study. Clin. Transl. Sci. 9, 168–175 (2016).

342

Shroff, G. & Gupta, R. Human embryonic stem cells in the treatment of patients with spinal cord injury. Ann. Neurosci. 22, 208–216 (2015).

343

Frantz, S. Embryonic stem cell pioneer Geron exits field, cuts losses. Nat. Biotechnol. 30, 12–13 (2012).

344

Li, J. Y., Christophersen, N. S., Hall, V., Soulet, D. & Brundin, P. Critical issues of clinical human embryonic stem cell therapy for brain repair. Trends Neurosci. 31, 146–153 (2008).

345

Rapalino, O. et al. Implantation of stimulated homologous macrophages results in partial recovery of paraplegic rats. Nat. Med. 4, 814–821 (1998).

346

Shin, T., Ahn, M., Moon, C., Kim, S. & Sim, K. B. Alternatively activated macrophages in spinal cord injury and remission: another mechanism for repair? Mol. Neurobiol. 47, 1011–1019 (2013).

347

Li, Z. et al. viaM2 macrophages promote PDGFRβ Pericytes Migration After Spinal Cord Injury in Mice PDGFB/PDGFRβ pathway. Front. Pharmacol. 12, 670813 (2021).

348

Knoller, N. et al. Clinical experience using incubated autologous macrophages as a treatment for complete spinal cord injury: phase Ⅰ study results. J. Neurosurg. Spine 3, 173–181 (2005).

349

Nazari, B. et al. miR-219 overexpressing oligodendrocyte progenitor cells for treating compression spinal cord injury. Metab. Brain Dis. 36, 1069–1077 (2021).

350

Duncan, G. J. et al. The fate and function of oligodendrocyte progenitor cells after traumatic spinal cord injury. Glia 68, 227–245 (2020).

351

Keirstead, H. S. et al. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J. Neurosci. 25, 4694–4705 (2005).

352

Kim, J. B. et al. Oct4-induced oligodendrocyte progenitor cells enhance functional recovery in spinal cord injury model. EMBO J. 34, 2971–2983 (2015).

353

Wang, Y., Piao, J. H., Larsen, E. C., Kondo, Y. & Duncan, I. D. Migration and remyelination by oligodendrocyte progenitor cells transplanted adjacent to focal areas of spinal cord inflammation. J. Neurosci. Res. 89, 1737–1746 (2011).

354

Manley, N., Priest, C., Denham, J., Wirth, E. & Lebkowski, J. Human embryonic stem cell-derived oligodendrocyte progenitor cells: preclinical efficacy and safety in cervical spinal cord injury. Stem Cells Transl. Med. 6, 1917–1929 (2017).

355

Zavvarian, M.-M., Toossi, A., Khazaei, M., Hong, J. & Fehlings, M. Novel innovations in cell and gene therapies for spinal cord injury. F1000Res 9, F1000 Faculty Rev-279 (2020).

356

Leng, Z.-K. et al. Cultivation, screening, identification and transplantation of Muse cell from human umbilical cord-derived for spinal cord injury in rats. Zhongguo Gu Shang 32, 327–334 (2019).

357

Fandel, T. M. et al. Transplanted human stem cell-derived interneuron precursors mitigate mouse bladder dysfunction and central neuropathic pain after spinal cord injury. Cell Stem Cell 19, 544–557 (2016).

358

Blesch, A. Human ESC-derived interneurons improve major consequences of spinal cord injury. Cell Stem Cell 19, 423–424 (2016).

359

Huang, J. H. et al. Systemic administration of exosomes released from mesenchymal stromal cells attenuates apoptosis, inflammation, and promotes angiogenesis after spinal cord injury in rats. J. Neurotrauma 34, 3388–3396 (2017).

360

Zhao, C. et al. Exosomes derived from bone marrow mesenchymal stem cells inhibit complement activation in rats with spinal cord injury. Drug Des. Devel. Ther. 13, 3693–3704 (2019).

361

Zhang, C. et al. Exosomes derived from human placenta-derived mesenchymal stem cells improve neurologic function by promoting angiogenesis after spinal cord injury. Neurosci. Lett. 739, 135399 (2020).

362

Frohlich, D. et al. Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 369, 20130510 (2014).

363

Jiang, D. et al. Neuron-derived exosomes-transmitted miR-124-3p protect traumatically injured spinal cord by suppressing the activation of neurotoxic microglia and astrocytes. J. Nanobiotechnol. 18, 105 (2020).

364

Shao, M. et al. Exosomes from long noncoding RNA-Gm37494-ADSCs repair spinal cord injury via shifting microglial M1/M2 polarization. Inflammation 43, 1536–1547 (2020).

365

Yuan, B., Pan, S., Dong, Y. Q., Zhang, W. W. & He, X. D. Effect of exosomes derived from mir-126-modified mesenchymal stem cells on the repair process of spinal cord injury in rats. Eur. Rev. Med. Pharmacol. Sci. 24, 4058 (2020).

366

Guo, S. et al. Intranasal delivery of mesenchymal stem cell derived exosomes loaded with phosphatase and tensin homolog siRNA repairs complete spinal cord injury. ACS Nano 13, 10015–10028 (2019).

367

Li, L. et al. Transplantation of human mesenchymal stem-cell-derived exosomes immobilized in an adhesive hydrogel for effective treatment of spinal cord injury. Nano Lett. 20, 4298–4305 (2020).

368

Wei, Z. et al. Proteomics analysis of Schwann cell-derived exosomes: a novel therapeutic strategy for central nervous system injury. Mol. Cell Biochem. 457, 51–59 (2019).

369

Rao, J. S. et al. NT3-chitosan enables de novo regeneration and functional recovery in monkeys after spinal cord injury. Proc. Natl. Acad. Sci. USA 115, E5595–E5604 (2018).

370

Yang, Z. et al. NT3-chitosan elicits robust endogenous neurogenesis to enable functional recovery after spinal cord injury. Proc. Natl. Acad. Sci. USA 112, 13354–13359 (2015).

371

Zhou, X. et al. Polycaprolactone electrospun fiber scaffold loaded with iPSCs-NSCs and ASCs as a novel tissue engineering scaffold for the treatment of spinal cord injury. Int J. Nanomed. 13, 6265–6277 (2018).

372

Saracino, G., Cigognini, D., Silva, D., Caprini, A. & Gelain, F. Nanomaterials design and tests for neural tissue engineering. Chem. Soc. Rev. 42, 225–262 (2013).

373

Xu, Y. et al. Understanding the role of tissue-specific decellularized spinal cord matrix hydrogel for neural stem/progenitor cell microenvironment reconstruction and spinal cord injury. Biomaterials 268, 120596 (2021).

374

Lu, P. et al. Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell 150, 1264–1273 (2012).

375

Liu, X. et al. 3D bioprinted neural tissue constructs for spinal cord injury repair. Biomaterials 272, 120771 (2021).

376

Zhou, X. et al. Three-dimensional-bioprinted dopamine-based matrix for promoting neural regeneration. ACS Appl. Mater. Interfaces 10, 8993–9001 (2018).

377

Lee, S. J. et al. 3D printing nano conductive multi-walled carbon nanotube scaffolds for nerve regeneration. J. Neural Eng. 15, 016018 (2018).

378

Koffler, J. et al. Biomimetic 3D-printed scaffolds for spinal cord injury repair. Nat. Med. 25, 263–269 (2019).

379

Joung, D. et al. 3D printed stem-cell derived neural progenitors generate spinal cord scaffolds. Adv. Funct. Mater. 28, 1801850 (2018).

380

Liu, W. et al. A functional scaffold to promote the migration and neuronal differentiation of neural stem/progenitor cells for spinal cord injury repair. Biomaterials 243, 119941 (2020).

381

Li, X. et al. A collagen microchannel scaffold carrying paclitaxel-liposomes induces neuronal differentiation of neural stem cells through Wnt/β-catenin signaling for spinal cord injury repair. Biomaterials 183, 114–127 (2018).

382

Xiao, Z. et al. Significant improvement of acute complete spinal cord injury patients diagnosed by a combined criteria implanted with NeuroRegen scaffolds and mesenchymal stem cells. Cell Transplant. 27, 907–915 (2018).

383

Duan, H. et al. Transcriptome analyses reveal molecular mechanisms underlying functional recovery after spinal cord injury. Proc. Natl. Acad. Sci. USA 112, 13360–13365 (2015).

384

Theodore, N. et al. First human implantation of a bioresorbable polymer scaffold for acute traumatic spinal cord injury: a clinical pilot study for safety and feasibility. Neurosurgery 79, E305–E312 (2016).

385

Kim, K. D. et al. A study of probable benefit of a bioresorbable polymer scaffold for safety and neurological recovery in patients with complete thoracic spinal cord injury: 6-month results from the INSPIRE study. J. Neurosurg. Spine, 1–10. (2021).

386

Caiazzo, M. et al. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476, 224–227 (2011).

387

Heinrich, C. et al. Directing astroglia from the cerebral cortex into subtype specific functional neurons. PLoS Biol. 8, e1000373 (2010).

388

Su, Z., Niu, W., Liu, M. L., Zou, Y. & Zhang, C. L. In vivo conversion of astrocytes to neurons in the injured adult spinal cord. Nat. Commun. 5, 3338 (2014).

389

Heinrich, C. et al. Sox2-mediated conversion of NG2 glia into induced neurons in the injured adult cerebral cortex. Stem Cell Rep. 3, 1000–1014 (2014).

390

Son, E. et al. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell 9, 205–218 (2011).

391

Zarei-Kheirabadi, M., Hesaraki, M., Kiani, S. & Baharvand, H. In vivo conversion of rat astrocytes into neuronal cells through neural stem cells in injured spinal cord with a single zinc-finger transcription factor. Stem Cell Res. Ther. 10, 380 (2019).

392

Matsuda, T. et al. Pioneer factor NeuroD1 rearranges transcriptional and epigenetic profiles to execute microglia-neuron conversion. Neuron 101, 472–485.e477 (2019).

393

Karow, M. et al. Direct pericyte-to-neuron reprogramming via unfolding of a neural stem cell-like program. Nat. Neurosci. 21, 932–940 (2018).

394

Hu, W. et al. Direct conversion of normal and Alzheimer’s DIsease Human Fibroblasts Into Neuronal Cells by Small Molecules. Cell Stem Cell 17, 204–212 (2015).

395

Chavez, A. et al. Highly efficient Cas9-mediated transcriptional programming. Nat. Methods 12, 326–328 (2015).

396

Wang, L. L. et al. Revisiting astrocyte to neuron conversion with lineage tracing in vivo. Cell 184, 5465–5481.e5416 (2021).

397

Patel, N. P. & Huang, J. H. Hyperbaric oxygen therapy of spinal cord injury. Med. Gas. Res. 7, 133–143 (2017).

398

Zhou, Y. et al. Hyperbaric oxygen improves functional recovery of the injured spinal cord by inhibiting inflammation and glial scar formation. Am. J. Phys. Med. Rehabil. 98, 914–920 (2019).

399

Huang, G. et al. Signaling pathways involved in HSP32 induction by hyperbaric oxygen in rat spinal neurons. Redox Biol. 10, 108–118 (2016).

400

Tan, J. W., Zhang, F., Liu, H. J. & Li, Z. Hyperbaric oxygen ameliorated the lesion scope and nerve function in acute spinal cord injury patients: a retrospective study. Clin. Biochem. 53, 1–7 (2018).

401

Sun, L. et al. Effect of hyperbaric oxygen therapy on HMGB1/NF-κB expression and prognosis of acute spinal cord injury: a randomized clinical trial. Neurosci. Lett. 692, 47–52 (2019).

402

Hwang, D. H., Park, H. H., Shin, H. Y., Cui, Y. & Kim, B. G. Insulin-like growth factor-1 receptor dictates beneficial effects of treadmill training by regulating survival and migration of neural stem cell grafts in the injured spinal cord. Exp. Neurobiol. 27, 489–507 (2018).

403

Hwang, D. H. et al. Survival of neural stem cell grafts in the lesioned spinal cord is enhanced by a combination of treadmill locomotor training via insulin-like growth factor-1 signaling. J. Neurosci. 34, 12788–12800 (2014).

404

Behrman, A. L., Ardolino, E. M. & Harkema, S. J. Activity-based therapy: from basic science to clinical application for recovery after spinal cord injury. J. Neurol. Phys. Ther. 41, S39–S45 (2017).

405

Shaffer, R. F., Picard, G. & Taylor, J. A. Relationship of spinal cord injury level and duration to peak aerobic capacity with arms-only and hybrid functional electrical stimulation rowing. Am. J. Phys. Med. Rehabil. 97, 488–491 (2018).

406

Zhang, C. et al. Early applied electric field stimulation attenuates secondary apoptotic responses and exerts neuroprotective effects in acute spinal cord injury of rats. Neuroscience 291, 260–271 (2015).

407

Li, G. et al. Epidural spinal cord stimulation promotes motor functional recovery by enhancing oligodendrocyte survival and differentiation and by protecting myelin after spinal cord injury in rats. Neurosci. Bull. 36, 372–384 (2020).

408

Hutson, T. H. & Di Giovanni, S. The translational landscape in spinal cord injury: focus on neuroplasticity and regeneration. Nat. Rev. Neurol. 15, 732–745 (2019).

409

Mishra, A. M., Pal, A., Gupta, D. & Carmel, J. B. Paired motor cortex and cervical epidural electrical stimulation timed to converge in the spinal cord promotes lasting increases in motor responses. J. Physiol. 595, 6953–6968 (2017).

410

Formento, E. et al. Electrical spinal cord stimulation must preserve proprioception to enable locomotion in humans with spinal cord injury. Nat. Neurosci. 21, 1728–1741 (2018).

411

Wenger, N. et al. Spatiotemporal neuromodulation therapies engaging muscle synergies improve motor control after spinal cord injury. Nat. Med. 22, 138–145 (2016).

412

Capogrosso, M. et al. A brain-spine interface alleviating gait deficits after spinal cord injury in primates. Nature 539, 284–288 (2016).

413

Wagner, F. B. et al. Targeted neurotechnology restores walking in humans with spinal cord injury. Nature 563, 65–71 (2018).

414

Greiner, N. et al. Recruitment of upper-limb motoneurons with epidural electrical stimulation of the cervical spinal cord. Nat. Commun. 12, 435 (2021).

Bone Research
Article number: 35
Cite this article:
Fan B, Wei Z, Feng S. Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Research, 2022, 10: 35. https://doi.org/10.1038/s41413-022-00199-9

151

Views

2

Downloads

84

Crossref

80

Web of Science

87

Scopus

Altmetrics

Received: 03 April 2021
Revised: 14 November 2021
Accepted: 22 December 2021
Published: 08 April 2022
© The Author(s) 2022

This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Return