Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1245 word(s) 1245 2021-07-13 11:24:46 |
2 format correct Meta information modification 1245 2021-07-14 05:45:37 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Suzuki, H. Chronic Spinal Cord Injury. Encyclopedia. Available online: https://encyclopedia.pub/entry/12057 (accessed on 29 March 2024).
Suzuki H. Chronic Spinal Cord Injury. Encyclopedia. Available at: https://encyclopedia.pub/entry/12057. Accessed March 29, 2024.
Suzuki, Hidenori. "Chronic Spinal Cord Injury" Encyclopedia, https://encyclopedia.pub/entry/12057 (accessed March 29, 2024).
Suzuki, H. (2021, July 14). Chronic Spinal Cord Injury. In Encyclopedia. https://encyclopedia.pub/entry/12057
Suzuki, Hidenori. "Chronic Spinal Cord Injury." Encyclopedia. Web. 14 July, 2021.
Chronic Spinal Cord Injury
Edit

Spinal cord injuries (SCI) are a devastating event and can lead to physical, psychosocial, and vocational implications for patients and their family. In the United States, approximately 288,000 individuals are estimated to suffer from symptoms caused by SCI, and a recent survey showed the annual incidence of SCI to be approximately 54 cases per one million people.

chronic spinal cord injury stem cells glial scar chondroitin sulfate proteoglycans regenerative medicine clinical trial

1. Overview

Chronic spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in clinical trials. In addition, several more are coming down the translational pipeline. Among ongoing and completed trials are those reporting the use of mesenchymal stem cells, neural stem/progenitor cells, induced pluripotent stem cells, olfactory ensheathing cells, and Schwann cells. The advancements in stem cell technology, combined with the powerful neuroimaging modalities, can now accelerate the pathway of promising novel therapeutic strategies from bench to bedside. Various combinations of different molecular therapies have been combined with supportive scaffolds to facilitate favorable cell–material interactions. In this review, we summarized some of the most recent insights into the preclinical and clinical studies using stem cells and other supportive drugs to unlock the microenvironment in chronic SCI to treat patients with this condition. Successful future therapies will require these stem cells and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, loss of structural framework, and immunorejection. 

2. Spinal Cord Injuries

Spinal cord injuries (SCI) are a devastating event and can lead to physical, psychosocial, and vocational implications for patients and their family. In the United States, approximately 288,000 individuals are estimated to suffer from symptoms caused by SCI, and a recent survey showed the annual incidence of SCI to be approximately 54 cases per one million people [1][2]. Worldwide, the estimated incidence of SCI ranges from 250,000–500,000 individuals per year [3]. The majority of neuroregenerative therapy has focused on treating patients in the acute or subacute periods. In the acute to subacute phase, salvageable neuronal cells may still exist and the glial scar has not yet been established [4][5][6][7]. Unfortunately, 95% of patients with SCI are in the chronic phase of their injury [8].
Despite this pressing need, one of the greatest challenges in developing an effective therapy for chronic SCI has been the inhibitory microenvironment of the injured spinal cord. After SCI, astrocytes activate, proliferate, and migrate to the perilesional region where they form a dense interwoven network of processes and deposit chondroitin sulfate proteoglycans (CSPGs) into the extracellular matrix. Dystrophic axons surround the injury epicenter and become trapped in the dense meshwork of scar tissue [9].
Various cell populations can be used for the treatment of chronic SCI. Concurrently, several clinical trials using stem cells are underway around the world [ClinicalTrials.gov. Available online: https://www.clinicaltrials.gov/ (accessed on 1 June 2021)]. Among them, exogenous neural stem cell (NSC) therapies are particularly promising as these cells have the potential to differentiate into all three neuroglial lineages (i.e., neurons, astrocytes, and oligodendrocytes) to regenerate neural circuits, remyelinate denuded axons, and provide trophic support to endogenous cells [7][9][10][11][12].

3. Barriers to Regeneration and Pathophysiology of Chronic SCI

It is widely recognized that regeneration of the adult mammalian central nervous system (CNS), including the spinal cord, is difficult due to its limited plasticity [13]. In the epicenter of a CNS lesion, a cavitation occurs that is surrounded by connective scar tissues containing cerebrospinal fluid. The phenotype of reactive astrocytes changes into scar-forming astrocytes that impede regenerating axons from crossing the lesion. Some inflammatory immune cells remain around the lesion even in the chronic phase of SCI (Figure 1).
Figure 1. The diagram shows the pathophysiological events in the chronic SCI. Injury to the SCI results in death of neuronal as well as glial cells. Progressive demyelination results in degeneration of axonal fibers that leads to disruption of axo-glial signaling. A cavitation has occurred in the epicenter. Hypertrophic astrocytes with very long processes over the tips of non-regenerating fibers form a barrier known as a glial barrier or a glial wall around the cavitation. In response to damage/injury, microglial cells transform into active phagocytic microglia and exhibit chemotaxis (migrates and accumulates at the site of injury). The presence of CSPGs creates an inhibitory environment for axonal regeneration, which leads to failure of axonal growth cones at the injured site of CNS. In addition, CSPG also inhibits the migration and differentiation of oligodendrocyte progenitor cells.

3.1. Astrocytic and Fibrotic Scar

Astrocytes proliferate and tightly interweave their extended processes around the perilesional region in an attempt to wall off the injury epicenter. Astrocytes, pericytes, and ependymal cells generate dense deposits of CSPGs as part of the fibrous scar, which bind leukocyte common antigen-related receptors such as protein tyrosine phosphatases. This activates GTPase RhoA and its downstream effector, rho-associated protein kinase (ROCK), leading to the collapse of the axonal growth cone and regenerative failure [14][15][16][17]. C3 transferase, an enzyme derived from Clostridium botulinum, locks RhoA in the inactive state and thereby inhibits Rho signaling. C3 transferase has been shown to promote axonal outgrowth on inhibitory substrates, both in vitro and in vivo [18][19]. The SPinal Cord Injury Rho INhibition InvestiGation (SPRING) clinical trial is now underway (NCT02669849) for acute SCI [2][20].

3.2. CSPGs and Chondroitinase ABC (ChABC)

CSPGs are a class of extracellular matrix molecule proteoglycans that are widely expressed within the CNS and that can be synthesized by all neural cell types [21]. CSPGs are highly upregulated in the glial scar after injury to the nervous system. In addition, they are mostly inhibitory and have been shown to hinder regeneration of axons across lesions in chronic SCI [22]. ChABC is a bacterial enzyme shown to effectively degrade CSPGs, including NG2, and to promote functional gains in mouse models after intrathecal administration [23][24]. Evidence also shows that co-administration of ChABC with neural precursor cells enhances transplant survival and remyelination of host axons [25][26]. More recently, large-scale CSPG digestion by direct lentiviral ChABC gene delivery into rat spinal cords resulted in a reduced cavitation volume and an enhanced preservation of axons. Treated rats also displayed an improved sensorimotor function on behavioral and electrophysiological assessments [27]. We also reported that ChABC administration reduced chronic injury scar and resulted in significantly improved NSCs derived from induced pluripotent stem cell (iPSC-NSC) survival with clear differentiation into all three neuroglial lineages. The chronically injured spinal cord can be ‘unlocked’ by ChABC pretreatment to produce a microenvironment conducive to regenerative iPSC therapy [9]. ChABC is an exciting therapy for which the optimal delivery modality remains to be elucidated. Future avenues of chronic SCI research may include exploration of human CNS-specific analogs of ChABC and its development.

4. Conclusions

Currently, numerous clinical and experimental studies have shown positive results in terms of functional improvement with stem cell treatment in chronic SCI. Various designs of chronic SCI trials need to be performed. However, human chronic SCI trials are not easily enforced because of some inherent limitations. First, comparison between treatment and control groups is difficult because of ethical aspects, and, in terms of safety and efficacy, the results of animal experiments cannot be directly applied to humans. Nevertheless, a lot of basic research and clinical trials of stem cell therapy have already been performed, and promising results have also been reported. We are convinced that stem cell therapy will provide the drastic treatment needed for chronic SCI patients in the near future.

References

  1. Fehlings, M.G.; Martin, A.R.; Tetreault, L.A.; Aarabi, B.; Anderson, P.; Arnold, P.M.; Brodke, D.S.; Burns, A.S.; Chiba, K.; Dettori, J.R.; et al. A clinical practice guideline for the management of patients with acute spinal cord injury: Recommendations on the role of baseline magnetic resonance imaging in clinical decision making and outcome prediction. Global Spine J. 2017, 7, 221s–230s.
  2. Fehlings, M.G.; Kim, K.D.; Aarabi, B.; Rizzo, M.; Lisa, M.B.; McKerracher, L.; Vaccaro, A.R.; David, O.; Okonkwo, D.O. Rho inhibitor VX-210 in acute traumatic subaxial cervical spinal cord injury: Design of the SPinal Cord Injury Rho Inhibition InvestiGation (SPRING) clinical trial. J. Neurotrauma 2018, 35, 1049–1056.
  3. Singh, A.; Tetreault, L.; Kalsi-Ryan, S.; Nouri, A.; Fehlings, M.G. Global prevalence and incidence of traumatic spinal cord injury. Clin. Epidemiol 2014, 6, 309–331.
  4. Ahuja, C.; Fehlings, M.G. Concise review: Bridging the gap: Novel neuroregenerative and neuroprotective strategies in spinal cord injury. Stem Cells Transl. Med. 2016, 5, 914–924.
  5. Zweckberger, K.; Ahuja, C.S.; Liu, Y.; Wang, J.; Fehlings, M.G. Self-assembling peptides optimize the post-traumatic milieu and synergistically enhance the effects of neural stem cell therapy after cervical spinal cord injury. Acta. Biomater. 2016, 42, 77–89.
  6. Kawabata, S.; Takano, M.; Numasawa-Kuroiwa, Y.; Itakura, G.; Kobayashi, Y.; Nishiyama, Y.; Sugai, K.; Nishimura, S.; Iwai, H.; Isoda, M.; et al. Grafted human iPS cell-derived oligodendrocyte precursor cells contribute to robust remyelination of demyelinated axons after spinal cord injury. Stem Cell Rep. 2016, 6, 1–8.
  7. Lu, P.; Kadoya, K.; Tuszynski, M.H. Axonal growth and connectivity from neural stem cell grafts in models of spinal cord injury. Curr. Opin. Neurobiol. 2014, 27, 103–109.
  8. Spinal cord injury (SCI) 2016 facts and figures at a glance. J. Spinal Cord Med. 2016, 9, 493–494.
  9. Suzuki, H.; Ahuja, C.S.; Salewski, R.P.; Li, L.; Satkunendrarajah, K.; Nagoshi, N.; Shibata, S.; Fehlings, M.G. Neural stem cell mediated recovery is enhanced by Chondroitinase ABC pretreatment in chronic cervical spinal cord injury. PLoS ONE 2017, 12, e0182339.
  10. Karimi-Abdolrezaee, S.; Eftekharpour, E.; Wang, J.; Morshead, C.M.; Fehlings, M.G. Delayed transplantation of adult neural precursor cells promotes remyelination and functional neurological recovery after spinal cord injury. J. Neurosci. 2006, 26, 3377–3389.
  11. Salewski, R.P.; Mitchell, R.A.; Shen, C.; Fehlings, M.G. Transplantation of neural stem cells clonally derived from embryonic stem cells promotes recovery after murine spinal cord injury. Stem Cells Dev. 2015, 24, 36–50.
  12. Wilcox, J.T.; Satkunendrarajah, K.; Zuccato, J.A.; Nassiri, F.; Fehlings, M.G. Neural precursor cell transplantation enhances functional recovery and reduces astrogliosis in bilateral compressive/contusive cervical spinal cord injury. Stem Cells Transl. Med. 2014, 3, 1148–1159.
  13. Ramon y Cajal, S. Degeneration and Regeneration of the Nervous System; Oxford University Press: London, UK, 1928.
  14. Cafferty, W.B.; Duffy, P.; Huebner, E.; Strittmatter, S.M. MAG and OMgp synergize with Nogo-A to restrict axonal growth and neurological recovery after spinal cord trauma. J. Neurosci. 2010, 30, 1825–1837.
  15. Chen, M.S.; Huber, A.B.; van der Haar, M.E.; Frank, M.; Schnell, L.; Spillmann, A.A.; Christ, F.; Schwab, M.E. Nogo-A is a myelin-associated neurite outgrowth inhibitor and an antigen for monoclonal antibody IN-1. Nature 2000, 403, 434–439.
  16. Forgione, N.; Fehlings, M.G. Rho-ROCK inhibition in the treatment of spinal cord injury. World Neurosurg. 2014, 82, e535–e539.
  17. Moreau-Fauvarque, C.; Kumanogoh, A.; Camand, E.; Jaillard, C.; Barbin, G.; Boquet, I.; Love, C.; Jones, E.Y.; Kikutani, H.; Lubetzki, C.; et al. The transmembrane semaphoring Sema4D/CD100, an inhibitor of axonal growth, is expressed on oligodendrocytes and upregulated after CNS lesion. J. Neurosci. 2003, 23, 9229–9239.
  18. Dubreuil, C.I.; Winton, M.J.; McKerracher, L. Rho activation patterns after spinal cord injury and the role of activated Rho in apoptosis in the central nervous system. J. Cell Biol. 2003, 162, 233–243.
  19. Lord-Fontaine, S.; Yang, F.; Diep, Q.; Dergham, P.; Munzer, S.; Tremblay, P.; McKerracher, L. Local inhibition of Rho signaling by cell-permeable recombinant protein BA-210 prevents secondary damage and promotes functional recovery following acute spinal cord injury. J. Neurotrauma 2008, 25, 1309–1322.
  20. Fehlings, M.G.; Theodore, N.; Harrop, J.; Maurais, G.; Kuntz, C.; Shaffrey, C.I.; Kwon, B.K.; Chapman, J.; Yee, A.; Tighe, A.; et al. A phase I/IIa clinical trial of a recombinant Rho protein antagonist in acute spinal cord injury. J. Neurotrauma 2011, 28, 787–796.
  21. Alizadeh, A.; Dyck, S.M.; Karimi-Abdolrezaee, S. Myelin damage and repair in pathologic CNS: Challenges and prospects. Front. Mol. Neurosci. 2015, 8, 35.
  22. Avram, S.; Shaposhnikov, S.; Buiu, C.; Mernea, M. Chondroitin sulfate proteoglycans: Structure-function relationship with implication in neural development and brain disorders. Biomed. Res. Int. 2014, 2014, 642798.
  23. Bradbury, E.J.; Moon, L.D.; Popat, R.J.; King, V.R.; Bennett, G.S.; Patel, P.N.; Fawcett, J.W.; McMahon, S.B. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 2002, 416, 636–640.
  24. Jones, L.L.; Sajed, D.; Tuszynski, M. Axonal regeneration through regions of chondroitin sulfate proteoglycan deposition after spinal cord injury: A balance of permissiveness and inhibition. J. Neurosci. 2003, 23, 9276–9288.
  25. Ikegami, T.; Nakamura, M.; Yamane, J.; Katoh, H.; Okada, S.; Iwanami, A.; Watanabe, K.; Ishii, K.; Kato, F.; Fujita, H.; 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.
  26. Carter, L.M.; Stephen, B.; McMahon, S.B.; Bradbury, E.J. Delayed treatment with chondroitinase ABC reverses chronic atrophy of rubrospinal neurons following spinal cord injury. Exp. Neurol. 2011, 228, 149–156.
  27. Bartus, K.; James, N.D.; Didangelos, A.; Bosch, K.D.; Verhaagen, J.; Yáñez-Muñoz, R.J.; Rogers, J.H.; Schneider, B.J.; Muir, E.M.; Bradbury, E.J. Large-scale chondroitin sulfate proteoglycan digestion with chondroitinase gene therapy leads to reduced pathology and modulates macrophage phenotype following spinal cord contusion injury. J. Neurosci. 2014, 34, 4822–4836.
More
Information
Subjects: Others
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 511
Revisions: 2 times (View History)
Update Date: 14 Jul 2021
1000/1000