Abstract
Spinal cord injury (SCI) leads to loss of nervous tissue and consequently to catastrophic neurological deficits. Up to now there is no definite treatment available that restores the loss of function to a degree that an independent life can be guaranteed.
This justifies the cost of research into the new modalities for a treatment of SCIs. In current paper, recent developments and new approaches in pharmacological therapy have been reviewed
Keywords: Medication therapy management, Spinal cord injury
Spinal cord injury (SCI) results in devastating neurological deficits and disabilities. The annual incidence of SCI in developed countries varies from 11.5 to 53.4 per million populations (1–3). As the life expectancy of survivors is increasing, information on the prevalence of SCI is becoming more important, but few prevalence studies are available.
Prevalence rate ranged from 11 to 112 per 100000 populations (4–7).
The social and economic impact of SCI extends to involve, not just the patient and the immediate family, but also the community and society at large (8, 9).
In 1990, it was estimated that the cost of managing all SCIs in the United States was 4 billion dollars annually.
Therefore, it seems rational to invest for the development of new treatment modalities that improve the condition of these patients.(10).
PATHOPHYSIOLOGY
There are two types of injury that may cause damage to the spinal cord. These include primary and secondary injuries (8). Primary injuries occur at the time of mechanical trauma to the spinal cord. Secondary injuries are caused by a series of cellular and biochemical reactions that are provoked by the primary injuries (11). Among these are hypoxia, accumulation of excitatory neurotransmitters, hyperthermia, imbalance in intracellular electrolytes, increased level of opioid (dynorphins)at the site of injury, free radical formation and lipid peroxidation, microvascular alteration, anemia, activation of inflammatory cells associated with cytokins and activation of calpain and caspaces and apoptosis (12, 13).
Inhibitory elements in the CNS inhibit damaged nerve fibers to exhibit regenerative sprouting. Nogo-A (neurite out growth inhibitor), MAG( myelin-associated glycoprotein), OMgp (oligodendrocyte-myelin glycoprotein), chondroitin sulfate proteoglycan which are exposed after the injury, are proteins that play such a role(14–16).
APPROACH TO TRAETMENT
Surgical decompression of the cord and restoration of its normal alignment, together with stabilization of the spine when indicated, accompanied by physical and occupation therapy, and managing psychologic, urologic and proctologic problems are the standard managing methods for spinal cord injury patients (17, 18).
Several attempts have been made to modulate the mechanisms leading to the secondary injury. These are: pharmacological interventions, neutralization of the inhibitory proteins, cell therapy, and gene therapy (19–22).
Considering the key pathophysiological mechanisms that contribute to neurological deficits after SCI, it seems that promising drug–based therapeutic approaches, (that we explain in this paper), including regenerative strategies to neutralize myelin-mediated neurite outgrowth inhibition, neuroprotective strategies to reduce apoptotic triggers, targeting of cationic/glutamatergic toxicity, anti-inflammatory strategies and the use of approaches that stabilize disrupted cell membranes could be considered effective ways to prevent further damage to the spinal cord and promote its repair(23, 24).
PHARMACOLOGICAL INTERVENTION
Methylprednisolone
Methylprednisolon (MP) has been used to treat different neurological impairments. Its mechanism of action has been explained by anti inflammatory and antioxidant properties in addition to inhibitory effects on lipid peroxidation (25). It has been suggested that MP selectively inhibits oligodendrocytes but not neuronal cell death via a receptor-mediated action and may be a mechanism for its limited protective effect after SCI (26). MP has been used together with olfactory ensheathing cells and Nogo-A monoclonal antibody after producing SCI in rat models; and improvement in axonal re-growth and neuroprotection have been observed(27, 28).
Efficacy has also been examined in a multi-centre double blind RCT by comparing high doses of MP (1000mg bolus and 1000mg daily thereafter for 10 days) with that of a standard dose (100mg bolus and 100 mg daily thereafter for 10 days). No significant difference was observed in neurological recovery 1 year after SCI between the two treatment groups (29). There is also no evidence of the benefit of its simultaneous use with nimodipine in the SCI (30).
Atorvastatin
Anti inflammatory properties and immunomodulatory activities of statins have been reported in the animal model previously (31). Atorvastatin treatment attenuated the SCI-induced inducible nitric oxide synthase (inos), tumor necrosis factor alpha (TNF-alpha), and interleukin 1beta (IL-beta) expression. Atorvastatin also provided protection against SCI-induced tissue necrosis, neuronal and oligodendrocyte apoptosis, demyelination, and reactive gliosis. Studies report beneficial effect of atorvastatin for the treatment of SCI- related pathology and disability.
Omega-3 Poly Unsaturated Fatty Acid (PUFA)
It has been suggested that PUFA could target some of the pathological mechanisms that underlie damage afterSCI (32). Reports indicated that omega-3 PUFA α-linolenic acid and docosahexaenoic acid (DHA) injected 30 min after injury significantly improved locomotor performance and neuroprotection, including a decrease in lesion size and apoptosis and an increase in neuronal and oligodendrocyte survival. In contrast, animals treated with arachidonic acid (AA), and omega-6 PUFA, had a significantly worse outcome than controls (33).
Fenretinide
Fenretinide (4-hydroxy (phenyl)retinamide) (INN) is a synthetic retinoid derivative. Retinoides are substances related to vitamin A, and they have been investigated for potential use in the treatment of cancer (34). Researches have shown that daily oral administration of fenretinide after spinal cord contusion injury led to a significant decrease in AA and increase in DHA levels in plasma and in the injured spinal cord tissue. This was accompanied by a significant reduction in tissue damage and improvement in locomotor recovery. Fenretinide also reduces the expression of proinflammatory genes and the levels of oxidative stress markers after SCI. In addition, vitro studies demonstrated that fenretinide reduces TNF-α expression by reactive microglia (35). These beneficial effects may be mediated via the ability of fenretinide to modulate PUFA hemostasis. Since fenretinide is currently in clinical trials for the treatment of cancers, this drug might be a good candidate for the treatment of acute SCI in humans.
Erythropoietin
Cytokine erythropoietin (EPO) is a glycoprotein mediating cytoprotection in a variety of tissues, including brain and spinal cord, through activation of multiple signaling pathways (36, 37). Uninjured spinal cord expresses a high density of EPO receptor (EPOR) in the basal state (38). It has been reported that EPO exerts its beneficial effects by apoptosis blockage, reduction of inflammation, and restoration of vascular integrity. Neuronal regeneration has also been suggested (39). Moreover, administration of EPO or other recently generated EPO analogues such as asialo-EPO and carbamilated-EPO demonstrate exceptional preclinical characteristics, rendering the evaluation of this tissue-protective agents imperative in human clinical trials (40).
Progestron
Reports show that progestron (Prog) has neuroprotective and promyelinating effects in lesions of the spinal cord(41). These effects may be due to regulation of myelin synthesis in glial cells and also to direct actions on neuronal function (42). When SCI is produced, several genes become sensitive to Prog. in the region caudal to the lesion site.
Neurotrophins, their receptors, and signaling cascades might be part of the molecules involved in the process of neuroprotection. It has been shown that in rats with SCI, a 3-day course of Prog. treatment increases the mRNA of brain-derived neurotrophic factor(BDNF)and BDNF immunoreactivity in perikaryon and processes of motoneurons, whereas chromotolysis is strongly prevented(43). Increased expression of BDNF is correlated with increased immunoreactivity of the BDNF receptor TrkB and of phosphorelated CAMP responsive element binding in motoneurons. In the same SCI model, Prog. restored myelination, according to measurements of myelin basic protein (MBP) and mRNA levels, and further increased the density of NG2+positive oligodendrocyte progenitors. These cells might be involved in remyelination of the damaged spinal cord.
Estrogen
Estrogen is a steroid that processes antiinflammatory and antioxidant effects (44). It may also modulate intracellular Ca(2+), attenuate apoptosis, and inhibit activation and activity of calpain and caspase-3(45, 46).
Treatment of SCI rats with estrogen (4mg/kg) at 15min and 24hr post-injury reduced edema and decreased inflammation and myelin loss in the lesion, suggesting its potential as a therapeutic agent (47).
Inosine
Inosine, a naturally occurring metabolite has been shown to induce axon outgrowth from primary neurons in culture through a direct intracellular mechanism(48). It can significantly reduce the spread of secondary
degeneration and the cell death following SCI(48, 49). Inosine applied with a minipump to the rat sensorimotor cortex stimulated intact pyramidal cells to undergo extensive sprouting of their axons into the denervated spinal cord white matter and adjacent neuropil (50, 51). Thus, inosine, a purin nucleoside without known side effects, might help to restore essential circuitry after injury to the CNS.
Rho Inhibitors (Cethrin-BA210)
Axons of the adult CNS have limited ability to regenerate after injury. This may be, at least partly, attributable to myelin-derived proteins, Nogo and oligodendrocyte myelin glycoprotein. Recent evidences suggest that these proteins inhibit neurit outgrowth by activation Rho (an intracellular GTPase) through the neurotrophin receptorP75NTR/Nogo receptor complex (52). Following SCI in vivo, massive activation of Rho is observed in the injured neurites (53–56). Application of Rho antagonist (c3-05) reversed Rho activation and reduced cell death after CNS injury. It also blocked the increase in P75NTR expression (57–59). Cethrine is a recombinant protein drug intended to facilitate the re-growth of axon during the critical period immediately after a major SCI (phase II trial is now going on). It can be delivered topically onto the dura matter (and diffuses into the spinal cord) during decompression/stabilization surgery (60, 61). Cethrin is known as an orphan drug by FDA.
cAMP-Rolipram
Elevation of intracellular cyclic AMP (C AMP) is one of the most successful strategies that enable axons to suppress inhibitory proteins that are made after SCI (62, 63). Intracellular cAMP levels can be increased through a peripheral conditioning lesion, administration of cAMP analogues, priming with neurotrophins or treatment with the phosphodistrase inhibitor rolipram (64).
Fingolimod (FTY720)
FTY720, a sphingosine receptor modulator that sequesters lymphocytes in secondary lymphoid organs, has been shown to be effective in the treatment of a variety of experimental autoimmune disorders (65, 66). Accordingly, by reducing lymphocyte infiltration into the spinal cord following SCI, this novel immunomodulator may enhance tissue preservation and functional recovery(67). Collectively, results demonstrate the neuroprotective potential of FTY720 after experimental SCI (68).
Chondroitinase ABC (Ch ABC)
For successful regeneration, long injured axons must overcome their poor intrinsic growth potential as well as the inhibitory environment of the glial scar established around the lesion site (69). Recent animal studies have shown that ch ABC can enhance plasticity in the adult nervous system by cleaving inhibitory chondroitin sulfate proteoglycans (CSPGs) in the scar matrix (70, 71). There are evidences indicating that CSPGs have two faces in spinal cord repair. Inhibition of CSGPs synthesis immediately after injury, impairs functional motor recovery and increases tissue loss. It causes a dramatic effect on the spatial organization of the infiltrating myeloid cells around the lesion site, decreases insulin-like growth factor 1 (IGF-1) production bymicroglia/macrophages, and increases TNF-α levels. In contrast, delayed inhibition, allowing CSPGs synthesis during the first two days fallowing injury, with subsequent inhibition, improves recovery (72). The distinction between the beneficial role of CSPGs during the acute stage and its deleterious effect at later stages emphasizes the need to retain the endogenous potential of this molecule in repair by controlling its levels at different stages of post-injury repair.
Sialidase
MAG, one of the inhibitor proteins, binds to sialoglycans and other receptors on axons. MAG inhibition of axon outgrowth in some neurons is reversed by treatment with sialidase, an enzyme that hydrolyzes sialic acids and eliminates MAG-sialoglycan binding. Neurotrophin-induced TrK tyrosin kinase receptor activation and neuronal cell survival responses have been reported to be under the control of a membrane associated sialidase(73). Reports show that sialidase delivered intrathecally to rats following SCI significantly enhances hindlimb motofunction, improves autonomic reflexes and increases axon sprouting (74, 75).
Calpain Inhibitor
Calpains are a family of calcium-dependent, non-lysosomal cystine proteases (proteolytic enzymes) (76, 77). One factor potentially limiting the efficacy of transplanted Schwann cells is poor cell survival. It has been demonstrated that SCs die within 24 hours after transplantation. Calpain inhibitor, MDL28170, enhances Schwann cell survival both in vitro in response to oxidative stress and in vivo following delayed transplantation into the injured spinal cord (78). The results support the use of calpain inhibitors as a promising new treatment for promoting the survival of transplanted cells79.
Scutellaria Baicalensis (EESB)
EESB significantly inhibits lipopolysacharide-induced expression of inflammatory mediators such as TNF-α, IL-1β, il-6, cycloxygenase-2, and inducible nitric oxide synthase. Additionally, reactive oxygen species and nitric oxide production are also significantly attenuated. In vivo study on rats with SCI that had orally received EESB, it was revealed that expression of proinflammatory factors and protein carbonylation and nitration were inhibited (80). Furthermore, EESB significantly inhibited apoptotic cell death of neurons and oligodendrocytes and improved functional recovery after SCI.
Folic acid
Reports demonstrate a crucial role for folate in the regeneration of afferent spinal neurons after injury. Findings show that axonal regeneration relies upon the integrity of DNA methylation pathways (81). It has been found that intraperitoneal treatment of adult rats with folic acid significantly improves the re-growth of sensory spinal axons into a grafted segment of peripheral nerve in vivo. The results show that the effects of folic acid supplementation on CNS growth processes are not restricted to the emberyonic period, but can also be effective for enhancing growth, repair, and recovery in injured adults CNS (82).
Pioglitazone
Peroxisome proliferation activated receptors (PPAR) are widely expressed nuclear receptors whose activation have led to diminished pro-inflammatory cascades in several CNS disorders. Based on this fact, efficacy of the PPAR agonist pioglitazone injected I.P in a rudent SCI model has been examined. Stereological measurement throughout the lesion revealed a significant increase in rostral spared white matter in pioglitazone treatment groups. Spinal cord from the high dose group also had significantly more gray matter sparing and motor neurons rostral and caudal to epicenter (83). These results suggest that clinical treatment with pioglitazone, an FDAapproved drug used currently for diabetes, may be a feasible and promising strategy for promoting anatomical and functional repair after SCI (84).
Anti- CD11d Integrin Antibody
Acute inflammatory response is a process that damages descending pathways after SCI. Neuropathic pain and motor dysfunction associated with SCI may be due to increases in serotonergic fiber density in spinal segments rostral and decreases caudal to the lesion. Restricting the acute inflammatory response by a monoclonal antibody to the CD11d subunit of the leukocyte CD11d/CD18 integrine, limits the trafficking of neutrophils and macrophages into the SCI site. After treatment, the typically increased serotonin immunoreactivity rostral to injury was reduced, whereas that caudal to the lesion increased toward normal. Increased serotonergic fiber density below the lesion also occurred in the intermediolateral cell column and ventral horn of treated rats, relative to controls. Improved locomotor recovery paralleled this increased serotonin (85, 86).
Nigella Sativa (Black Seed)
Nigella sativa (NS) is a plant, native to southwest Asia. It grows to 20-30 cm tall, with finely divided, linear leaves. NS has been used for medicinal proposes for centuries, as a herb and it was also pressed into oil, in Asia, middle East and Africa (87–89). Possible beneficial effects of NS have been investigated on experimental SCI in rats. Malondialdehyde (MDA) and protein carbonylation (PC) levels are increased in SCI, whereas, superoxide dismutase (SOD), glutathion peroxidase (GSH-PX), and catalase (CAT) enzyme activities decreased. NS treatment decreased tissue MDA and PC levels and prevented inhibition of SOD, GSH-PX and CAT enzymes in the tissue. The morphology of neurons in NS-treated groups was well protected. As a result, NS treatment might be helpful in SCI and therefore shows a potential for clinical implication (90–92).
Sodium Channel Blockers (Riluzole, Phenytoin, Mexiletine)
Influx of Na (+) ions into cells have been postulated to be a key early event in the pathogenesis of secondary traumatic and ischemic CNS injury. Pharmacological blockade of Na (+) channels can attenuate secondary pathophysiology and reduce functional deficits acutely. In an experimental model of rats with SCI riluzole, injected I.P significantly enhanced residual tissue area at the injury epicenter compared with controls. Riluzole (a drug used to treat ALS, preferentially blocks tetrodotoxin sensitive sodium channels, which are associated with damaged neurons) (93, 94)significantly reduced tissue loss in rostro caudal regions surrounding the epicenter, with overall sparing of gray matter and selective sparing of white matter(95, 96).
Animal experiments with phenytoin also showed decreased MDA levels in rats with SCI. Phenytoin appears to protect spinal cord against injury by decreasing lipidperoxidation and lessening neural damage associated with SCI in rats(97–99). Some studies reveal better results with riluzole and mexiletine than phenytoin (95, 100).
Hydralazine (Acrolein Scavenger Drug)
Acrolein, a lipid peroxidation byproduct, is significantly increased following SCI in vivo, and that exposure to neuronal cells results in oxidative stress, mitochondrial dysfunction, increased membrane permeability, impaired axonal conductivity and eventually cell death (101). Acrolein thus may be a key player in the pathogenesis of SCI, where lipid peroxidation is known to be involved (102). Acrolein is capable of depleting endogenous antioxidants such as glutathione, generating free radicals, and damaging proteins and DNA (103).
Acrolein has a significantly longer half-life than the transient free radicals, and thus may represent a potentially better target of therapeutic intervention to attenuate oxidative stress. Animal studies demonstrate that the Acrolein scavenger hydralazine protects against not only Acrolein- mediated injury, but also compression in SCI. It can significantly alleviate Acrolein- induced superoxide production, glutathione depletion, mitochondrial dysfunction, loss of membrane integrity and reduced compound action potential conduction (104). These findings suggest that Acrolein scavenging drugs such as hydralazine may represent a novel therapy to effectively reduce oxidative stress in SCI.
Minocycline (An Antibiotic Drug)
After SCI, pro-nerve growh factor (proNGF) plays a central role in apoptosis of oligodendrocytes. It has been shown that minocycline improves functional recovery after SCI in part by reducing apoptosis of oligodendrocytes via inhibition of pro NGF production in macroglia (105). Minocycline treatment significantly reduces production in microglia in vitro and invivo by inhibition of the phosphorylation of p38MAKP (a protein kinase) (106). Minocycline treatment significantly reduces a number of terminal deoxinucleotidyle transferase (TdT)-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL)-positive cells 24h after SCI as compared to that of the vehicle control(107). In addition, minocycline treatment significantly reduces the specific caspase-3 activity after SCI. It also decreases TNF-α expression. Furthermore, minocycline treatment inhibits p75 neurotrophin receptor expression and Rho A activation after injury. These data suggest that after SCI, minocycline treatment modulated expression of cytokins, attenuated cell death and the size of lesion, and improved functional recovery in injured rats.
GHRELIN
Ghrelin, an acylated peptide, synthesized primarily in stomach and is the endogenouse ligand for the growth hormone secretagogue receptor (GHS-R), stimulates both food intake and GH secretion (108, 109). Ghrelin is not detected in normal, uninjured spinal cord, but spinal cord neurons and oligodendrocytes express the Ghrelin receptor (110). Ghrelin significantly inhibits apoptotic cell death of neurons and oligodendrocytes, release of mitochondrial cytochrome-C and activation of caspase-3 after moderate contusive SCI. Ghrelin also significantly increases the level of phosphorylated ERK (protein kinase) but decreases the level of phosphorylated p38MAPK. In addition, ghrelin increases the level of ERK-dependant brain-derived neurotrophic factor expression and decreases the level of pronerve growth factor expression. The neuroprotective effects of ghrelin are mediated through the ghrelin receptor. Eventually, ghrelin significantly improves functional recovery and reduces the size of lesion volume and the loss of axons and myelin after injury(111).
4-Aminopyridine (4-Ap) and Its Derivatives
4-AP is a potassium (k+) channel blocking agent that has been shown to reduce the latency and increases the amplitude of motor evoked potentials (MEPS) elicited with transcranial magnetic stimulation (TMS) in patients with chronic SCI (112). It has been shown to improve neurological motor and sensory function in both animal and human studies, and it has a more effect on conduction after stretch injury than compression (113, 114). Thedrug is safe, however, after starting 4-AP therapy, patients must be carefully monitored for the possible occurrence of peripheral vasospasm (which may be the most hazardous complication of this therapy) (115, 116).
Since the maximal tolerated level of 4-AP in humans is 100 times lower than the optimal dose in animal studies, structurally distinct pyridine based blocker with fewer side effects have been synthesized(117). One of these derivatives named (4-AP-3-MeOH) is 10 times more potent than 4-AP in restoring axonal conduction (118).
Conclusion
The complicated and sometimes confusing mechanisms of SCI had caused therapeutic interventions to improve very slowly for many years. However, in recent years, rapid progress that has occurred in understanding the pathophysiology of SCI, opened up the way for new pharmacologic therapies. These new strategies in addition to other treatment approaches (cell therapy, gene therapy, immunomodulatory therapy and bioscaffolds) have shown promising results to help patients with SCI.
References
- 1.Botterell EH, Jousse AT, Kraus AS, Thompson MG, WynneJones M, Geisler WO. A model for the future care of acute spinal cord injuries. Can J Neurol Sci. 1975;2:361–380. doi: 10.1017/s0317167100020497. [DOI] [PubMed] [Google Scholar]
- 2.Kraus JF, Silberman TA, McArthur DL. Epidemiology of spinal cord injury. In: Menezes AH, Sonntag VKH, editors. Principles of spinal surgery. NEW YORK: McGraw-Hill; 1996. [Google Scholar]
- 3.Van den Berg ME, Castellote JM, Mahillo-Fernandez I, de Pedro-Cuesta J. Incidence of spinal cord injury worldwide: a systematic review. Neuroepidemiology. 2010;34:184–192. doi: 10.1159/000279335. [DOI] [PubMed] [Google Scholar]
- 4.National Spinal Cord Injury Statistical Center. Spinal cord injury. facts and figures at a glance. J Spinal Cord Med. 2005;28:379–380. [PubMed] [Google Scholar]
- 5.Anderson DW, Kalsbeek WD. The National Head and Spinal Cord Injury Survey: assessment of some uncertainties affecting the findings. J Neurosurg. 1980;(Suppl):32–34. [PubMed] [Google Scholar]
- 6.Ergas Z. Spinal cord injury in the United States: a statistical update. Cent Nerv Syst Trauma. 1985;2:19–32. doi: 10.1089/cns.1985.2.19. [DOI] [PubMed] [Google Scholar]
- 7.Harvey C, Rothschild BB, Asmann AJ, Stripling T. New estimates of traumatic SCI prevalence: a survey-based approach. Paraplegia. 1990;28:537–544. doi: 10.1038/sc.1990.73. [DOI] [PubMed] [Google Scholar]
- 8.Sekhon LH, Fehlings MG. Epidemiology, demographics, and pathophysiology of acute spinal cord injury. Spine. 2001;26(24 suppl):S2–12. doi: 10.1097/00007632-200112151-00002. [DOI] [PubMed] [Google Scholar]
- 9.Berkowitz M, Harvey C, Greene CG, Wilson SE. Economic Consequences of Traumatic Spinal Cord Injury. N Engl J Med. 1994;330:1464. [Google Scholar]
- 10.Stripling TE. The cost of economic consequences of traumatic spinal cord injury. Paraplegia News. 1990;8:50–54. [Google Scholar]
- 11.David S, Lacroix S. Molecular approaches to spinal cord repair. Annu Rev Neurosci. 2003;26:411–440. doi: 10.1146/annurev.neuro.26.043002.094946. [DOI] [PubMed] [Google Scholar]
- 12.Abraham KE, Brewer KL, McGinty JF. Opioid peptide messenger RNA expression is increased at spinal and supraspinal levels following excitotoxic spinal cord injury. Neuroscience. 2000;99:189–197. doi: 10.1016/s0306-4522(00)00150-0. [DOI] [PubMed] [Google Scholar]
- 13.Abraham KE, McGinty JF, Brewer KL. The role of kainic acid/AMPA and metabotropic glutamate receptors in the regulation of opioid mRNA expression and the onset of pain-related behavior following excitotoxic spinal cord injury. Neuroscience. 2001;104:863–874. doi: 10.1016/s0306-4522(01)00134-8. [DOI] [PubMed] [Google Scholar]
- 14.Filbin MT. Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci. 2003;4:703–713. doi: 10.1038/nrn1195. [DOI] [PubMed] [Google Scholar]
- 15.Hulsebosch CE. Recent advances in pathophysiology and treatment of spinal cord injury. Adv Physiol Educ. 2002;26:238–255. doi: 10.1152/advan.00039.2002. [DOI] [PubMed] [Google Scholar]
- 16.Onose G, Anghelescu A, Muresanu DF, Padure L, Haras MA, Chendreanu CO, et al. A review of published reports on neuroprotection in spinal cord injury. Spinal Cord. 2009;47:716–726. doi: 10.1038/sc.2009.52. [DOI] [PubMed] [Google Scholar]
- 17.Silber JS, Vaccaro AR. Summary statement: the role and timing of decompression in acute spinal cord injury: evidence-based guidelines. Spine. 2001;26:S101–110. doi: 10.1097/00007632-200112151-00018. [DOI] [PubMed] [Google Scholar]
- 18.Fehlings MG, Sekhon LH, Tator C. The role and timing of decompression in acute spinal cord injury: what do we know? What should we do? Spine. 2001;26:S101–110. doi: 10.1097/00007632-200112151-00017. [DOI] [PubMed] [Google Scholar]
- 19.Sayer FT, Kronvall E, Nilsson OG. Methylprednisolone treatment in acute spinal cord injury: the myth challenged through a structured analysis of published literature. Spine. 2006;6:335–343. doi: 10.1016/j.spinee.2005.11.001. [DOI] [PubMed] [Google Scholar]
- 20.Baptiste DC, Fehlings MG. Pharmacological approaches to repair the injured spinal cord. J Neurotrauma. 2006;23:318–334. doi: 10.1089/neu.2006.23.318. [DOI] [PubMed] [Google Scholar]
- 21.Blits B, Bunge MB. Direct gene therapy for repair of the spinal cord. J Neurotrauma. 2006;23:508–520. doi: 10.1089/neu.2006.23.508. [DOI] [PubMed] [Google Scholar]
- 22.Pearse DD, Bunge MB. Designing cell- and gene-based regeneration strategies to repair the injured spinal cord. J Neurotrauma. 2006;23:438–452. doi: 10.1089/neu.2006.23.437. [DOI] [PubMed] [Google Scholar]
- 23.Scott AL, Ramer LM, Soril LJ, Kwiecien JM, Ramer MS. Targeting myelin to optimize plasticity of spared spinal axons. Mol Neurobiol. 2006;33:91–111. doi: 10.1385/MN:33:2:91. [DOI] [PubMed] [Google Scholar]
- 24.Baptiste DC, Fehlings MG. Emerging drugs for spinal cord injury. Expert Opin Emerg Drugs. 2008;13:63–80. doi: 10.1517/14728214.13.1.63. [DOI] [PubMed] [Google Scholar]
- 25.Bishnoi M, Chopra K, Kulkarni SK. U-74500A (lazaroid), a 21-aminosteroid attenuates neuroleptic-induced orofacial dyskinesia. Methods Find Exp Clin Pharmacol. 2007;29:601–605. doi: 10.1358/mf.2007.29.9.1161006. [DOI] [PubMed] [Google Scholar]
- 26.Lee JM, Yan P, Xiao Q, Chen S, Lee KY, Hsu CY, et al. Methylprednisolone protects oligodendrocytes but not neurons after spinal cord injury. J Neurosci. 2008;28:3141–3149. doi: 10.1523/JNEUROSCI.5547-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wu J, Yang H, Qiu Z, Zhang Q, Ding T, Geng D. Effect of combined treatment with methylprednisolone and Nogo-A monoclonal antibody after rat spinal cord injury. J Int Med Res. 2010;38:570–582. doi: 10.1177/147323001003800219. [DOI] [PubMed] [Google Scholar]
- 28.Nash HH, Borke RC, Anders JJ. Ensheathing cells and methylprednisolone promote axonal regeneration and functional recovery in the lesioned adult rat spinal cord. J Neurosci. 2002;22:7111–7120. doi: 10.1523/JNEUROSCI.22-16-07111.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bracken MB, Shepard MJ, Hellenbrand KG, Collins WF, Leo LS, Freeman DF, et al. Methylprednisolone and neurological function 1 year after spinal cord injury. Results of the National Acute Spinal Cord Injury Study. J Neurosurg. 1985;63:704–713. doi: 10.3171/jns.1985.63.5.0704. [DOI] [PubMed] [Google Scholar]
- 30.Petitjean ME, Pointillart V, Dixmerias F, Wiart L, Sztark F, Lassie P, et al. Medical treatment of spinal cord injury in the acute stage] Ann Fr Anesth Reanim. 1998;17:114–122. doi: 10.1016/s0750-7658(98)80058-0. [DOI] [PubMed] [Google Scholar]
- 31.Pannu R, Barbosa E, Singh AK, Singh I. Attenuation of acute inflammatory response by atorvastatin after spinal cord injury in rats. J Neurosci Res. 2005;79:340–350. doi: 10.1002/jnr.20345. [DOI] [PubMed] [Google Scholar]
- 32.Michael-Titus AT. Omega-3 fatty acids: their neuroprotective and regenerative potential in traumatic neurological injury. Clin Lipidology. 2009;4:343–353. [Google Scholar]
- 33.King VR, Huang WL, Dyall SC, Curran OE, Priestley JV, Michael-Titus AT. Omega-3 fatty acids improve recovery, whereas omega-6 fatty acids worsen outcome, after spinal cord injury in the adult rat. J Neurosci. 2006;26:4672–4680. doi: 10.1523/JNEUROSCI.5539-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wu JM, DiPietrantonio AM, Hsieh TC. Mechanism of fenretinide (4-HPR)-induced cell death. Apoptosis. 2001;6:377–388. doi: 10.1023/a:1011342220621. [DOI] [PubMed] [Google Scholar]
- 35.Lopez-Vales R, Redensek A, Skinner TA, Rathore KI, Ghasemlou N, Wojewodka G, et al. Fenretinide promotes functional recovery and tissue protection after spinal cord contusion injury in mice. J Neurosci. 2010;30:3220–3226. doi: 10.1523/JNEUROSCI.5770-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Byts N, Siren AL. Erythropoietin: a multimodal neuroprotective agent. Exp Transl Stroke Med. 2009;1:4. doi: 10.1186/2040-7378-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Siren AL, Fasshauer T, Bartels C, Ehrenreich H. Therapeutic potential of erythropoietin and its structural or functional variants in the nervous system. Neurotherapeutics. 2009;6:108–127. doi: 10.1016/j.nurt.2008.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Grasso G, Sfacteria A, Erbayraktar S, Passalacqua M, Meli F, Gokmen N, et al. Amelioration of spinal cord compressive injury by pharmacological preconditioning with erythropoietin and a nonerythropoietic erythropoietin derivative. J Neurosurg Spine. 2006;4:310–318. doi: 10.3171/spi.2006.4.4.310. [DOI] [PubMed] [Google Scholar]
- 39.Vitellaro-Zuccarello L, Mazzetti S, Madaschi L, Bosisio P, Gorio A, De Biasi S. Erythropoietin-mediated preservation of the white matter in rat spinal cord injury. Neuroscience. 2007;144:865–877. doi: 10.1016/j.neuroscience.2006.10.023. [DOI] [PubMed] [Google Scholar]
- 40.Matis GK, Birbilis TA. Erythropoietin in spinal cord injury. Eur Spine J. 2009;18:314–323. doi: 10.1007/s00586-008-0829-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.De Nicola AF, Gonzalez SL, Labombarda F, Deniselle MC, Garay L, Guennoun R, et al. Progesterone treatment of spinal cord injury: Effects on receptors, neurotrophins, and myelination. J Mol Neurosci. 2006;28:3–15. doi: 10.1385/jmn:28:1:3. [DOI] [PubMed] [Google Scholar]
- 42.Gonzalez SL, Labombarda F, Gonzalez Deniselle MC, Guennoun R, Schumacher M, De Nicola AF. Progesterone up-regulates neuronal brain-derived neurotrophic factor expression in the injured spinal cord. Neuroscience. 2004;125:605–614. doi: 10.1016/j.neuroscience.2004.02.024. [DOI] [PubMed] [Google Scholar]
- 43.Labombarda F, Gonzalez S, Gonzalez Deniselle MC, Garay L, Guennoun R, Schumacher M, et al. Progesterone increases the expression of myelin basic protein and the number of cells showing NG2 immunostaining in the lesioned spinal cord. J Neurotrauma. 2006;23:181–192. doi: 10.1089/neu.2006.23.181. [DOI] [PubMed] [Google Scholar]
- 44.Kachadroka S, Hall AM, Niedzielko TL, Chongthammakun S, Floyd CL. Effect of endogenous androgens on 17beta-estradiol-mediated protection after spinal cord injury in male rats. J Neurotrauma. 2010;27:611–626. doi: 10.1089/neu.2009.1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sribnick EA, Samantaray S, Das A, Smith J, Matzelle DD, Ray SK, et al. Postinjury estrogen treatment of chronic spinal cord injury improves locomotor function in rats. J Neurosci Res. 2010;88:1738–1750. doi: 10.1002/jnr.22337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sribnick EA, Matzelle DD, Ray SK, Banik NL. Estrogen treatment of spinal cord injury attenuates calpain activation and apoptosis. J Neurosci Res. 2006;84:1064–1075. doi: 10.1002/jnr.21016. [DOI] [PubMed] [Google Scholar]
- 47.Ritz MF, Hausmann ON. Effect of 17beta-estradiol on functional outcome, release of cytokines, astrocyte reactivity and inflammatory spreading after spinal cord injury in male rats. Brain Res. 2008;1203:177–188. doi: 10.1016/j.brainres.2008.01.091. [DOI] [PubMed] [Google Scholar]
- 48.Liu F, You SW, Yao LP, Liu HL, Jiao XY, Shi M, et al. Secondary degeneration reduced by inosine after spinal cord injury in rats. Spinal Cord. 2006;44:421–426. doi: 10.1038/sj.sc.3101878. [DOI] [PubMed] [Google Scholar]
- 49.Conta AC, Stelzner DJ. Immunomodulatory effect of the purine nucleoside inosine following spinal cord contusion injury in rat. Spinal Cord. 2008;46:39–44. doi: 10.1038/sj.sc.3102057. [DOI] [PubMed] [Google Scholar]
- 50.Benowitz LI, Goldberg DE, Madsen JR, Soni D, Irwin N. Inosine stimulates extensive axon collateral growth in the rat corticospinal tract after injury. Proc Natl Acad Sci U S A. 1999;96:13486–13490. doi: 10.1073/pnas.96.23.13486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zai L, Ferrari C, Subbaiah S, Havton LA, Coppola G, Strittmatter S, et al. Inosine alters gene expression and axonal projections in neurons contralateral to a cortical infarct and improves skilled use of the impaired limb. J Neurosci. 2009;29:8187–8197. doi: 10.1523/JNEUROSCI.0414-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bhakar AL, Howell JL, Paul CE, Salehi AH, Becker EB, Said F, et al. Apoptosis induced by p75NTR overexpression requires Jun kinase-dependent phosphorylation of Bad. J Neurosci. 2003;23:11373–11381. doi: 10.1523/JNEUROSCI.23-36-11373.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Madura T, Yamashita T, Kubo T, Fujitani M, Hosokawa K, Tohyama M. Activation of Rho in the injured axons following spinal cord injury. EMBO Rep. 2004;5:412–417. doi: 10.1038/sj.embor.7400117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.McKerracher L, Higuchi H. Targeting Rho to stimulate repair after spinal cord injury. J Neurotrauma. 2006;23:309–317. doi: 10.1089/neu.2006.23.309. [DOI] [PubMed] [Google Scholar]
- 55.Scott AL, Ramer MS. Schwann cell p75NTR prevents spontaneous sensory reinnervation of the adult spinal cord. Brain. 2010;133:421–432. doi: 10.1093/brain/awp316. [DOI] [PubMed] [Google Scholar]
- 56.Baptiste DC, Tighe A, Fehlings MG. Spinal cord injury and neural repair: focus on neuroregenerative approaches for spinal cord injury. Expert Opin Investig Drugs. 2009;18:663–673. doi: 10.1517/13543780902897623. [DOI] [PubMed] [Google Scholar]
- 57.Dubreuil CI, Winton MJ, 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. doi: 10.1083/jcb.200301080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Ellezam B, Dubreuil C, Winton M, Loy L, Dergham P, Selles-Navarro I, et al. Inactivation of intracellular Rho to stimulate axon growth and regeneration. Prog Brain Res. 2002;137:371–380. doi: 10.1016/s0079-6123(02)37028-6. [DOI] [PubMed] [Google Scholar]
- 59.Cosgaya JM, Chan JR, Shooter EM. The neurotrophin receptor p75NTR as a positive modulator of myelination. Science. 2002;298:1245–1248. doi: 10.1126/science.1076595. [DOI] [PubMed] [Google Scholar]
- 60.Dergham P, Ellezam B, Essagian C, Avedissian H, Lubell WD, McKerracher L. Rho signaling pathway targeted to promote spinal cord repair. J Neurosci. 2002;22:6570–6577. doi: 10.1523/JNEUROSCI.22-15-06570.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lord-Fontaine S, Yang F, Diep Q, Dergham P, Munzer S, Tremblay P, et al. 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. doi: 10.1089/neu.2008.0613. [DOI] [PubMed] [Google Scholar]
- 62.Neumann S, Bradke F, Tessier-Lavigne M, Basbaum AI. Regeneration of sensory axons within the injured spinal cord induced by intraganglionic cAMP elevation. Neuron. 2002;34:885–893. doi: 10.1016/s0896-6273(02)00702-x. [DOI] [PubMed] [Google Scholar]
- 63.Qiu J, Cai D, Dai H, McAtee M, Hoffman PN, Bregman BS, et al. Spinal axon regeneration induced by elevation of cyclic AMP. Neuron. 2002;34:895–903. doi: 10.1016/s0896-6273(02)00730-4. [DOI] [PubMed] [Google Scholar]
- 64.Hannila SS, Filbin MT. The role of cyclic AMP signaling in promoting axonal regeneration after spinal cord injury. Exp Neurol. 2008;209:321–332. doi: 10.1016/j.expneurol.2007.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Paugh SW, Payne SG, Barbour SE, Milstien S, Spiegel S. The immunosuppressant FTY720 is phosphorylated by sphingosine kinase type 2. FEBS Lett. 2003;554:189–193. doi: 10.1016/s0014-5793(03)01168-2. [DOI] [PubMed] [Google Scholar]
- 66.Billich A, Bornancin F, Devay P, Mechtcheriakova D, Urtz N, Baumruker T. Phosphorylation of the immunomodulatory drug FTY720 by sphingosine kinases. J Biol Chem. 2003;278:47408–47415. doi: 10.1074/jbc.M307687200. [DOI] [PubMed] [Google Scholar]
- 67.Horga A, Montalban X. FTY720 (fingolimod) for relapsing multiple sclerosis. Expert Rev Neurother. 2008;8:699–714. doi: 10.1586/14737175.8.5.699. [DOI] [PubMed] [Google Scholar]
- 68.Lee KD, Chow WN, Sato-Bigbee C, Graf MR, Graham RS, Colello RJ, et al. FTY720 reduces inflammation and promotes functional recovery after spinal cord injury. J Neurotrauma. 2009;26:2335–2344. doi: 10.1089/neu.2008.0840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tom VJ, Sandrow-Feinberg HR, Miller K, Santi L, Connors T, Lemay MA, et al. Combining peripheral nerve grafts and chondroitinase promotes functional axonal regeneration in the chronically injured spinal cord. J Neurosci. 2009;29:14881–14890. doi: 10.1523/JNEUROSCI.3641-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jakeman LB, Hoschouer EL, Basso DM. Injured mice at the gym: Review, results and considerations for combining chondroitinase and locomotor exercise to enhance recovery after spinal cord injury. Brain Res Bull; 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bai F, Peng H, Etlinger JD, Zeman RJ. Partial functional recovery after complete spinal cord transection by combined chondroitinase and clenbuterol treatment. Pflugers Arch. 2010;460:657–666. doi: 10.1007/s00424-010-0852-y. [DOI] [PubMed] [Google Scholar]
- 72.Rolls A, Shechter R, London A, Segev Y, Jacob-Hirsch J, Amariglio N, et al. Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med. 2008;5:e171. doi: 10.1371/journal.pmed.0050171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Jayanth P, Amith SR, Gee K, Szewczuk MR. Neu1 sialidase and matrix metalloproteinase-9 cross-talk is essential for neurotrophin activation of Trk receptors and cellular signaling. Cell Signal. 2010;22:1193–1205. doi: 10.1016/j.cellsig.2010.03.011. [DOI] [PubMed] [Google Scholar]
- 74.Mountney A, Zahner MR, Lorenzini I, Oudega M, Schramm LP, Schnaar RL. Sialidase enhances recovery from spinal cord contusion injury. Proc Natl Acad Sci U S A. 2010;107:11561–11566. doi: 10.1073/pnas.1006683107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yang LJ, Lorenzini I, Vajn K, Mountney A, Schramm LP, Schnaar RL. Sialidase enhances spinal axon outgrowth in vivo. Proc Natl Acad Sci U S A. 2006;103:11057–11062. doi: 10.1073/pnas.0604613103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ohno S, Emori Y, Imajoh S, Kawasaki H, Kisaragi M, Suzuki K. Evolutionary origin of a calcium-dependent protease by fusion of genes for a thiol protease and a calcium-binding protein? Nature. 1984;312:566–570. doi: 10.1038/312566a0. [DOI] [PubMed] [Google Scholar]
- 77.Goll DE, Thompson VF, Li H, Wei W, Cong J. The calpain system. Physiol Rev. 2003;83:731–801. doi: 10.1152/physrev.00029.2002. [DOI] [PubMed] [Google Scholar]
- 78.Hill CE, Guller Y, Raffa SJ, Hurtado A, Bunge MB. A calpain inhibitor enhances the survival of schwann cells in vitro and after transplantation into the injured spinal cord. J Neurotrauma. 2010;27:1685–1695. doi: 10.1089/neu.2010.1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Xiong Y, Hall ED. Pharmacological evidence for a role of peroxynitrite in the pathophysiology of spinal cord injury. Exp Neurol. 2009;216:105–114. doi: 10.1016/j.expneurol.2008.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Yune TY, Lee JY, Cui CM, Kim HC, Oh TH. Neuroprotective effect of Scutellaria baicalensis on spinal cord injury in rats. J Neurochem. 2009;110:1276–1287. doi: 10.1111/j.1471-4159.2009.06214.x. [DOI] [PubMed] [Google Scholar]
- 81.Kronenberg G, Endres M. Neuronal injury: folate to the rescue? J Clin Invest. 2010;120:1383–1386. doi: 10.1172/JCI40764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Iskandar BJ, Nelson A, Resnick D, Skene JH, Gao P, Johnson C, et al. Folic acid supplementation enhances repair of the adult central nervous system. Ann Neurol. 2004;56:221–227. doi: 10.1002/ana.20174. [DOI] [PubMed] [Google Scholar]
- 83.McTigue DM, Tripathi R, Wei P, Lash AT. The PPAR gamma agonist Pioglitazone improves anatomical and locomotor recovery after rodent spinal cord injury. Exp Neurol. 2007;205:396–406. doi: 10.1016/j.expneurol.2007.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Smith U. Pioglitazone: mechanism of action. Int J Clin Pract. 2001;(Suppl):13–18. [PubMed] [Google Scholar]
- 85.Oatway MA, Chen Y, Bruce JC, Dekaban GA, Weaver LC. Anti-CD11d integrin antibody treatment restores normal serotonergic projections to the dorsal, intermediate, and ventral horns of the injured spinal cord. J Neurosci. 2005;25:637–647. doi: 10.1523/JNEUROSCI.3960-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bao F, Dekaban GA, Weaver LC. Anti-CD11d antibody treatment reduces free radical formation and cell death in the injured spinal cord of rats. J Neurochem. 2005;94:1361–1373. doi: 10.1111/j.1471-4159.2005.03280.x. [DOI] [PubMed] [Google Scholar]
- 87.Ghonime M, Eldomany R, Abdelaziz A, Soliman H. Evaluation of immunomodulatory effect of three herbal plants growing in Egypt. Immunopharmacol Immunotoxicol; 2010. [DOI] [PubMed] [Google Scholar]
- 88.Hajhashemi V, Ghannadi A, Jafarabadi H. Black cumin seed essential oil, as a potent analgesic and antiinflammatory drug. Phytother Res. 2004;18:195–199. doi: 10.1002/ptr.1390. [DOI] [PubMed] [Google Scholar]
- 89.Lupidi G, Scire A, Camaioni E, Khalife KH, De Sanctis G, Tanfani F, et al. Thymoquinone, a potential therapeutic agent of Nigella sativa, binds to site I of human serum albumin. Phytomedicine. 2010;17:714–720. doi: 10.1016/j.phymed.2010.01.011. [DOI] [PubMed] [Google Scholar]
- 90.Kanter M, Coskun O, Kalayci M, Buyukbas S, Cagavi F. Neuroprotective effects of Nigella sativa on experimental spinal cord injury in rats. Hum Exp Toxicol. 2006;25:127–133. doi: 10.1191/0960327106ht608oa. [DOI] [PubMed] [Google Scholar]
- 91.El-Naggar T, Gomez-Serranillos MP, Palomino OM, Arce C, Carretero ME. Nigella sativa L. seed extract modulates the neurotransmitter amino acids release in cultured neurons in vitro. J Biomed Biotechnol. 2010;2010:398312. doi: 10.1155/2010/398312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Mousavi SH, Tayarani-Najaran Z, Asghari M, Sadeghnia HR. Protective effect of Nigella sativa extract and thymoquinone on serum/glucose deprivation-induced PC12 cells death. Cell Mol Neurobiol. 2010;30:591–598. doi: 10.1007/s10571-009-9484-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Song JH, Huang CS, Nagata K, Yeh JZ, Narahashi T. Differential action of riluzole on tetrodotoxin-sensitive and tetrodotoxin-resistant sodium channels. J Pharmacol Exp Ther. 1997;282:707–714. [PubMed] [Google Scholar]
- 94.Kitzman PH. Effectiveness of riluzole in suppressing spasticity in the spinal cord injured rat. Neurosci Lett. 2009;455:150–153. doi: 10.1016/j.neulet.2009.03.016. [DOI] [PubMed] [Google Scholar]
- 95.Schwartz G, Fehlings MG. Evaluation of the neuroprotective effects of sodium channel blockers after spinal cord injury: improved behavioral and neuroanatomical recovery with riluzole. J Neurosurg. 2001;94:245–256. doi: 10.3171/spi.2001.94.2.0245. [DOI] [PubMed] [Google Scholar]
- 96.Ates O, Cayli SR, Gurses I, Karabulut AB, Yucel N, Kocak A, et al. Do sodium channel blockers have neuroprotective effect after onset of ischemic insult? Neurol Res. 2007;29:317–323. doi: 10.1179/016164107X159225. [DOI] [PubMed] [Google Scholar]
- 97.Kaptanoglu E, Solaroglu I, Surucu HS, Akbiyik F, Beskonakli E. Blockade of sodium channels by phenytoin protects ultrastructure and attenuates lipid peroxidation in experimental spinal cord injury. Acta Neurochir. 2005;147:405–412. doi: 10.1007/s00701-004-0447-5. [DOI] [PubMed] [Google Scholar]
- 98.Schwartz G, Fehlings MG. Secondary injury mechanisms of spinal cord trauma: a novel therapeutic approach for the management of secondary pathophysiology with the sodium channel blocker riluzole. Prog Brain Res. 2002;137:177–190. doi: 10.1016/s0079-6123(02)37016-x. [DOI] [PubMed] [Google Scholar]
- 99.Mu X, Azbill RD, Springer JE. Riluzole improves measures of oxidative stress following traumatic spinal cord injury. Brain Res. 2000;870:66–72. doi: 10.1016/s0006-8993(00)02402-1. [DOI] [PubMed] [Google Scholar]
- 100.Ates O, Cayli SR, Gurses I, Turkoz Y, Tarim O, Cakir CO, et al. Comparative neuroprotective effect of sodium channel blockers after experimental spinal cord injury. J Clin Neurosci. 2007;14:658–665. doi: 10.1016/j.jocn.2006.03.023. [DOI] [PubMed] [Google Scholar]
- 101.Burcham PC, Kaminskas LM, Tan D, Pyke SM. Carbonyl-scavenging drugs & protection against carbonyl stress-associated cell injury. Mini Rev Med Chem. 2008;8:319–330. doi: 10.2174/138955708783955953. [DOI] [PubMed] [Google Scholar]
- 102.Hamann K, Durkes A, Ouyang H, Uchida K, Pond A, Shi R. Critical role of acrolein in secondary injury following ex vivo spinal cord trauma. J Neurochem. 2008;107:712–721. doi: 10.1111/j.1471-4159.2008.05622.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hamann K, Shi R. Acrolein scavenging: a potential novel mechanism of attenuating oxidative stress following spinal cord injury. J Neurochem. 2009;111:1348–1356. doi: 10.1111/j.1471-4159.2009.06395.x. [DOI] [PubMed] [Google Scholar]
- 104.Hamann K, Nehrt G, Ouyang H, Duerstock B, Shi R. Hydralazine inhibits compression and acrolein-mediated injuries in ex vivo spinal cord. J Neurochem. 2008;104:708–718. doi: 10.1111/j.1471-4159.2007.05002.x. [DOI] [PubMed] [Google Scholar]
- 105.Stirling DP, Khodarahmi K, Liu J, McPhail LT, McBride CB, Steeves JD, et al. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J Neurosci. 2004;24:2182–2190. doi: 10.1523/JNEUROSCI.5275-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Yune TY, Lee JY, Jung GY, Kim SJ, Jiang MH, Kim YC, et al. Minocycline alleviates death of oligodendrocytes by inhibiting pro-nerve growth factor production in microglia after spinal cord injury. J Neurosci. 2007;27:7751–7761. doi: 10.1523/JNEUROSCI.1661-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lee SM, Yune TY, Kim SJ, Park DW, Lee YK, Kim YC, et al. Minocycline reduces cell death and improves functional recovery after traumatic spinal cord injury in the rat. J Neurotrauma. 2003;20:1017–1027. doi: 10.1089/089771503770195867. [DOI] [PubMed] [Google Scholar]
- 108.Lim CT, Kola B, Korbonits M, Grossman AB. Ghrelin's role as a major regulator of appetite and its other functions in neuroendocrinology. Prog Brain Res. 2010;182:189–205. doi: 10.1016/S0079-6123(10)82008-4. [DOI] [PubMed] [Google Scholar]
- 109.O'Brien M, Earley P, Morrison JJ, Smith TJ. Ghrelin in the human myometrium. Reprod Biol Endocrinol. 2010;8:55. doi: 10.1186/1477-7827-8-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Mondal MS, Date Y, Yamaguchi H, Toshinai K, Tsuruta T, Kangawa K, et al. Identification of ghrelin and its receptor in neurons of the rat arcuate nucleus. Regul Pept. 2005;126:55–59. doi: 10.1016/j.regpep.2004.08.038. [DOI] [PubMed] [Google Scholar]
- 111.Lee JY, Chung H, Yoo YS, Oh YJ, Oh TH, Park S, et al. Inhibition of apoptotic cell death by ghrelin improves functional recovery after spinal cord injury. Endocrinology. 2010;151:3815–3826. doi: 10.1210/en.2009-1416. [DOI] [PubMed] [Google Scholar]
- 112.Wolfe DL, Hayes KC, Hsieh JT, Potter PJ. Effects of 4-aminopyridine on motor evoked potentials in patients with spinal cord injury: a double-blinded, placebo-controlled crossover trial. J Neurotrauma. 2001;18:757–771. doi: 10.1089/089771501316919120. [DOI] [PubMed] [Google Scholar]
- 113.Jensen JM, Shi R. Effects of 4-aminopyridine on stretched mammalian spinal cord: the role of potassium channels in axonal conduction. J Neurophysiol. 2003;90:2334–2340. doi: 10.1152/jn.00868.2002. [DOI] [PubMed] [Google Scholar]
- 114.Segal JL, Brunnemann SR. 4-Aminopyridine alters gait characteristics and enhances locomotion in spinal cord injured humans. J Spinal Cord Med. 1998;21:200–204. doi: 10.1080/10790268.1998.11719527. [DOI] [PubMed] [Google Scholar]
- 115.Grijalva I, Guizar-Sahagun G, Castaneda-Hernandez G, Mino D, Maldonado-Julian H, Vidal-Cantu G, et al. Efficacy and safety of 4-aminopyridine in patients with long-term spinal cord injury: a randomized, double-blind, placebo-controlled trial. Pharmacotherapy. 2003;23:823–834. doi: 10.1592/phco.23.7.823.32731. [DOI] [PubMed] [Google Scholar]
- 116.Segal JL, Pathak MS, Hernandez JP, Himber PL, Brunnemann SR, Charter RS. Safety and efficacy of 4-aminopyridine in humans with spinal cord injury: a long-term, controlled trial. Pharmacotherapy. 1999;19:713–723. doi: 10.1592/phco.19.9.713.31540. [DOI] [PubMed] [Google Scholar]
- 117.McBride JM, Smith DT, Byrn SR, Borgens RB, Shi R. 4-Aminopyridine derivatives enhance impulse conduction in guinea-pig spinal cord following traumatic injury. Neuroscience. 2007;148:44–52. doi: 10.1016/j.neuroscience.2007.05.039. [DOI] [PubMed] [Google Scholar]
- 118.Sun W, Smith D, Fu Y, Cheng JX, Bryn S, Borgens R, et al. Novel potassium channel blocker, 4-AP-3-MeOH, inhibits fast potassium channels and restores axonal conduction in injured guinea pig spinal cord white matter. J Neurophysiol. 2010;103:469–478. doi: 10.1152/jn.00154.2009. [DOI] [PubMed] [Google Scholar]