Skip to main content
Top
Published in: Journal of Orthopaedic Surgery and Research 1/2017

Open Access 01-12-2017 | Research article

Effect of oxidative stress induced by intracranial iron overload on central pain after spinal cord injury

Authors: Fan Xing Meng, Jing Ming Hou, Tian Sheng Sun

Published in: Journal of Orthopaedic Surgery and Research | Issue 1/2017

Login to get access

Abstract

Background

Central pain (CP) is a common clinical problem in patients with spinal cord injury (SCI). Recent studies found the pathogenesis of CP was related to the remodeling of the brain. We investigate the roles of iron overload and subsequent oxidative stress in the remodeling of the brain after SCI.

Methods

We established a rat model of central pain after SCI. Rats were divided randomly into four groups: SCI, sham operation, SCI plus deferoxamine (DFX) intervention, and SCI plus nitric oxide synthase (NOS) inhibitor treatment. Pain behavior was observed and thermal pain threshold was measured regularly, and brain levels of iron, transferrin receptor 1 (TfR1), ferritin (Fn), and lactoferrin (Lf), were detected in the different groups 12 weeks after establishment of the model.

Results

Rats demonstrated self-biting behavior after SCI. Furthermore, the latent period of thermal pain was reduced and iron levels in the hind limb sensory area, hippocampus, and thalamus increased after SCI. Iron-regulatory protein (IRP) 1 levels increased in the hind limb sensory area, while Fn levels decreased. TfR1 mRNA levels were also increased and oxidative stress was activated. Oxidative stress could be inhibited by ferric iron chelators and NOS inhibitors.

Conclusions

SCI may cause intracranial iron overload through the NOS–iron-responsive element/IRP pathway, resulting in central pain mediated by the oxidative stress response. Iron chelators and oxidative stress inhibitors can effectively relieve SCI-associated central pain.
Literature
1.
go back to reference Bonica JJ. History of pain concepts and pain therapy. Mt Sinai J Med. 1991;58(3):191–202.PubMed Bonica JJ. History of pain concepts and pain therapy. Mt Sinai J Med. 1991;58(3):191–202.PubMed
3.
go back to reference Yezierski RP. Pain following spinal cord injury: the clinical problem and experimental studies. Pain. 1996;68(2–3):185–94. Review.CrossRefPubMed Yezierski RP. Pain following spinal cord injury: the clinical problem and experimental studies. Pain. 1996;68(2–3):185–94. Review.CrossRefPubMed
4.
go back to reference Klega A, Eberle T, Buchholz HG, et al. Central opioidergic neurotransmission in complex regional pain syndrome. Neurology. 2010;75(2):129–36.CrossRefPubMed Klega A, Eberle T, Buchholz HG, et al. Central opioidergic neurotransmission in complex regional pain syndrome. Neurology. 2010;75(2):129–36.CrossRefPubMed
5.
go back to reference Baastrup C, Finnerup NB. Pharmacological management of neuropathic pain following spinal cord injury. CNS Drugs. 2008;22(6):455–75.CrossRefPubMed Baastrup C, Finnerup NB. Pharmacological management of neuropathic pain following spinal cord injury. CNS Drugs. 2008;22(6):455–75.CrossRefPubMed
6.
go back to reference Buckalew N, Haut MW, Morrow L, et al. Chronic pain is associated with brain volume loss in older adults: preliminary evidence. Pain Med. 2008;9(2):240–8.CrossRefPubMed Buckalew N, Haut MW, Morrow L, et al. Chronic pain is associated with brain volume loss in older adults: preliminary evidence. Pain Med. 2008;9(2):240–8.CrossRefPubMed
7.
go back to reference Gustin SM, Wrigley PJ, Siddall PJ, et al. Brain anatomy changes associated with persistent neuropathic pain following spinal cord injury. Cereb Cortex. 2010;20(6):1409–19.CrossRefPubMed Gustin SM, Wrigley PJ, Siddall PJ, et al. Brain anatomy changes associated with persistent neuropathic pain following spinal cord injury. Cereb Cortex. 2010;20(6):1409–19.CrossRefPubMed
8.
go back to reference Likavcanova K, Urdzikova L, Hajek M, et al. Metabolic changes in the thalamus after spinal cord injury followed by proton MR spectroscopy. Magn Reson Med. 2008;59(3):499–506.CrossRefPubMed Likavcanova K, Urdzikova L, Hajek M, et al. Metabolic changes in the thalamus after spinal cord injury followed by proton MR spectroscopy. Magn Reson Med. 2008;59(3):499–506.CrossRefPubMed
9.
go back to reference Jin L, Wang J, Zhao L, et al. Decreased serum ceruloplasmin levels characteristically aggravate nigral iron deposition in Parkinson’s disease. Brain. 2011;134(Pt 1):50–8.CrossRefPubMed Jin L, Wang J, Zhao L, et al. Decreased serum ceruloplasmin levels characteristically aggravate nigral iron deposition in Parkinson’s disease. Brain. 2011;134(Pt 1):50–8.CrossRefPubMed
10.
go back to reference Chen Z, Gao C, Hua Y, et al. Role of iron in brain injury after intraventricular hemorrhage. Stroke. 2011;42(2):465–70.CrossRefPubMed Chen Z, Gao C, Hua Y, et al. Role of iron in brain injury after intraventricular hemorrhage. Stroke. 2011;42(2):465–70.CrossRefPubMed
11.
go back to reference Duce JA, Tsatsanis A, Cater MA, et al. Iron-export ferroxidase activity of beta-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell. 2010;142(6):857–67.CrossRefPubMedPubMedCentral Duce JA, Tsatsanis A, Cater MA, et al. Iron-export ferroxidase activity of beta-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell. 2010;142(6):857–67.CrossRefPubMedPubMedCentral
12.
go back to reference Hadzhieva M, Kirches E, Wilisch-Neumann A, et al. Dysregulation of iron protein expression in the G93A model of amyotrophic lateral sclerosis. Neuroscience. 2013;230(6):94–101.CrossRefPubMed Hadzhieva M, Kirches E, Wilisch-Neumann A, et al. Dysregulation of iron protein expression in the G93A model of amyotrophic lateral sclerosis. Neuroscience. 2013;230(6):94–101.CrossRefPubMed
13.
go back to reference Gaasch JA, Lockman PR, Geldenhuys WJ, et al. Brain iron toxicity: differential responses of astrocytes neurons and endothelial cells. Neurochem Res. 2007;32(7):1196–208.CrossRefPubMed Gaasch JA, Lockman PR, Geldenhuys WJ, et al. Brain iron toxicity: differential responses of astrocytes neurons and endothelial cells. Neurochem Res. 2007;32(7):1196–208.CrossRefPubMed
14.
go back to reference Chen CW, Chen QB, Ouyang Q, et al. Transient early neurotrophin release and delayed inflammatory cytokine release by microglia in response to PAR-2 stimulation. J Neuroinflammation. 2012;9:142.PubMedPubMedCentral Chen CW, Chen QB, Ouyang Q, et al. Transient early neurotrophin release and delayed inflammatory cytokine release by microglia in response to PAR-2 stimulation. J Neuroinflammation. 2012;9:142.PubMedPubMedCentral
15.
go back to reference Smith JA, Das A, Ray SK, et al. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87(1):10–20.CrossRefPubMed Smith JA, Das A, Ray SK, et al. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87(1):10–20.CrossRefPubMed
16.
go back to reference Davidof G, Roth E, Guarracini M, et al. Function limiting dysesthetic pain syndrome among traumatic SCI patients: a cross sectional study. Pain. 1987;29:39–48.CrossRef Davidof G, Roth E, Guarracini M, et al. Function limiting dysesthetic pain syndrome among traumatic SCI patients: a cross sectional study. Pain. 1987;29:39–48.CrossRef
17.
19.
go back to reference Nepomuceno C, Fine PR, Richards JS, et al. Pain in patients with spinal cord injury. Arch Phys Med Rehabil. 1979;60(12):605–9.PubMed Nepomuceno C, Fine PR, Richards JS, et al. Pain in patients with spinal cord injury. Arch Phys Med Rehabil. 1979;60(12):605–9.PubMed
20.
go back to reference Miki K, Iwata K, Tsuboi Y, et al. Dorsal column-thalamic pathway is involved in thalamic hyperexcitability following peripheral nerve injury: a lesion study in rats with experimental mononeuropathy. Pain. 2000;85(1–2):263–71.CrossRefPubMed Miki K, Iwata K, Tsuboi Y, et al. Dorsal column-thalamic pathway is involved in thalamic hyperexcitability following peripheral nerve injury: a lesion study in rats with experimental mononeuropathy. Pain. 2000;85(1–2):263–71.CrossRefPubMed
21.
go back to reference Sokal DM, Chapman V. Effects of spinal administration of muscimol on C- and A-fibre evoked neuronal responses of spinal dorsal horn neurones in control and nerve injured rats. Brain Res. 2003;962(1–2):213–20.CrossRefPubMed Sokal DM, Chapman V. Effects of spinal administration of muscimol on C- and A-fibre evoked neuronal responses of spinal dorsal horn neurones in control and nerve injured rats. Brain Res. 2003;962(1–2):213–20.CrossRefPubMed
22.
go back to reference Dougherty PM, Palecek J, Paleckova V, et al. The role of NMDA and non-NMDA excitatory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, chemical, thermal, and electrical stimuli. Neurosci. 1992;12(8):3025–41. Dougherty PM, Palecek J, Paleckova V, et al. The role of NMDA and non-NMDA excitatory amino acid receptors in the excitation of primate spinothalamic tract neurons by mechanical, chemical, thermal, and electrical stimuli. Neurosci. 1992;12(8):3025–41.
23.
go back to reference Melzack R, Loeser JD. Phantom body pain in paraplegics: evidence for a central “pattern generating mechanism” for pain. Pain. 1978;4(3):195–210.PubMed Melzack R, Loeser JD. Phantom body pain in paraplegics: evidence for a central “pattern generating mechanism” for pain. Pain. 1978;4(3):195–210.PubMed
24.
go back to reference Evseev VA, Davydova TV, Vetrile LA. Common neuroimmunological features of drug addiction, alcoholism, epilepsy, and neurogenic pain syndromes. Vestn Ross Akad Med Nauk. 2006;2006(7):38–43. Evseev VA, Davydova TV, Vetrile LA. Common neuroimmunological features of drug addiction, alcoholism, epilepsy, and neurogenic pain syndromes. Vestn Ross Akad Med Nauk. 2006;2006(7):38–43.
25.
go back to reference Peyron R, Schneider F, Faillenot I, et al. An fMRI study of cortical representation of mechanical allodynia in patients with neuropathic pain. Neurology. 2004;63(10):1838–46.CrossRefPubMed Peyron R, Schneider F, Faillenot I, et al. An fMRI study of cortical representation of mechanical allodynia in patients with neuropathic pain. Neurology. 2004;63(10):1838–46.CrossRefPubMed
26.
go back to reference Wrigley PJ, Press SR, Gustin SM, et al. Neuropathic pain and primary somatosensory cortex reorganization following spinal cord injury. Pain. 2009;141(1–2):52–9.CrossRefPubMed Wrigley PJ, Press SR, Gustin SM, et al. Neuropathic pain and primary somatosensory cortex reorganization following spinal cord injury. Pain. 2009;141(1–2):52–9.CrossRefPubMed
27.
go back to reference Richards JS, Meredith RL, Nepomuceno C, et al. Psycho-social aspects of chronic pain in spinal cord injury. Pain. 1980;8(3):355–66.CrossRefPubMed Richards JS, Meredith RL, Nepomuceno C, et al. Psycho-social aspects of chronic pain in spinal cord injury. Pain. 1980;8(3):355–66.CrossRefPubMed
28.
go back to reference Summers JD, Rapoff MA, Varghese G, et al. Psychosocial factors in chronic spinal cord injury pain. Pain. 1991;47:183–9.CrossRefPubMed Summers JD, Rapoff MA, Varghese G, et al. Psychosocial factors in chronic spinal cord injury pain. Pain. 1991;47:183–9.CrossRefPubMed
29.
go back to reference Thompson FJ, Reier PJ, Lucas CC, Parmer R, et al. Altered patterns of reflex excitability subsequent to contusion injury of the rat spinal cord. J Neurophysiol. 1992;68(5):1473–86.PubMed Thompson FJ, Reier PJ, Lucas CC, Parmer R, et al. Altered patterns of reflex excitability subsequent to contusion injury of the rat spinal cord. J Neurophysiol. 1992;68(5):1473–86.PubMed
30.
go back to reference Dora CD, Koch S, Sanchez A, et al. Intraspinal injection of adenosine agonists protect against L-NAME induced neuronal loss in the rat. J Neurotrauma. 1998;15(7):473–83.CrossRefPubMed Dora CD, Koch S, Sanchez A, et al. Intraspinal injection of adenosine agonists protect against L-NAME induced neuronal loss in the rat. J Neurotrauma. 1998;15(7):473–83.CrossRefPubMed
31.
go back to reference Yezierski RP, Liu S, Ruenes GL, et al. Excitotoxicspinal cord injury:behavioral and morphological characteris-tics of a central pain model. Pain. 1998;75(1):141–55.CrossRefPubMed Yezierski RP, Liu S, Ruenes GL, et al. Excitotoxicspinal cord injury:behavioral and morphological characteris-tics of a central pain model. Pain. 1998;75(1):141–55.CrossRefPubMed
32.
go back to reference Thompson FJ, Reier PJ, Lucas CC, et al. Alteredpatterns of reflex excitability subsequent to contusion injuryof the rat spinal cord. J Neurophysiol. 1992;68(6):1473–86.PubMed Thompson FJ, Reier PJ, Lucas CC, et al. Alteredpatterns of reflex excitability subsequent to contusion injuryof the rat spinal cord. J Neurophysiol. 1992;68(6):1473–86.PubMed
33.
go back to reference Atwood CS, Obrenovich ME, Liu T, Chan H, Perry G, Smith MA, Martins RN. Amyloid-beta: a chameleon walking in two worlds: a review of the trophic and toxic properties of amyloid-beta. Brain Res Brain Res Rev. 2003;43(1):1–16.CrossRefPubMed Atwood CS, Obrenovich ME, Liu T, Chan H, Perry G, Smith MA, Martins RN. Amyloid-beta: a chameleon walking in two worlds: a review of the trophic and toxic properties of amyloid-beta. Brain Res Brain Res Rev. 2003;43(1):1–16.CrossRefPubMed
34.
36.
go back to reference Aisen P, et al. Iron metabolism. CurrOPinChemBiol. 1999;3:200–6. Aisen P, et al. Iron metabolism. CurrOPinChemBiol. 1999;3:200–6.
37.
go back to reference Jellinger KA. The role of iron in neuro degeneration. DrugsAging. 1999;14:115–14. Jellinger KA. The role of iron in neuro degeneration. DrugsAging. 1999;14:115–14.
38.
go back to reference Qian ZM, Wang Q. Expression of iron transport prote in sand excessiveiron accumulation of iron in the brain in neuro degenerative disorders. BrainResRev. 1998;27:257–67. Qian ZM, Wang Q. Expression of iron transport prote in sand excessiveiron accumulation of iron in the brain in neuro degenerative disorders. BrainResRev. 1998;27:257–67.
39.
go back to reference Wang L, Wang W, Zhao M, Ma L, Li M. Psychological stress induces dysregulation of iron metabolism in rat brain. Neuroscience. 2008;155(1):24–30.CrossRefPubMed Wang L, Wang W, Zhao M, Ma L, Li M. Psychological stress induces dysregulation of iron metabolism in rat brain. Neuroscience. 2008;155(1):24–30.CrossRefPubMed
40.
go back to reference Berg D, Youdim MB. Role of iron in neurodegenerative disorders. Top Magn Reson Imaging. 2006;17(1):5–17.CrossRefPubMed Berg D, Youdim MB. Role of iron in neurodegenerative disorders. Top Magn Reson Imaging. 2006;17(1):5–17.CrossRefPubMed
41.
42.
go back to reference Jalalvand E, Javan M, Haeri-Rohani A, Ahmadiani A. Stress- and non-stress-mediated mechanisms are involved in pain-induced apoptosis in hippocampus and dorsal lumbar spinal cord in rats. Neuroscience. 2008;157:446–52.CrossRefPubMed Jalalvand E, Javan M, Haeri-Rohani A, Ahmadiani A. Stress- and non-stress-mediated mechanisms are involved in pain-induced apoptosis in hippocampus and dorsal lumbar spinal cord in rats. Neuroscience. 2008;157:446–52.CrossRefPubMed
43.
go back to reference Vartiainen N, Kirveskari E, Kallio-Laine K, Kalso E, Forss N. Cortical reorganization in primary somatosensory cortex in patients with unilateral chronic pain. J Pain. 2009;10(8):854–9.CrossRefPubMed Vartiainen N, Kirveskari E, Kallio-Laine K, Kalso E, Forss N. Cortical reorganization in primary somatosensory cortex in patients with unilateral chronic pain. J Pain. 2009;10(8):854–9.CrossRefPubMed
44.
go back to reference Ji J, Zhou Y, Hao S, Wang Q. Low expression of ferroxidases is implicated in the iron retention in human atherosclerotic plaques. Biochem Biophys Res Commun. 2015;464(4):1134–8.CrossRefPubMed Ji J, Zhou Y, Hao S, Wang Q. Low expression of ferroxidases is implicated in the iron retention in human atherosclerotic plaques. Biochem Biophys Res Commun. 2015;464(4):1134–8.CrossRefPubMed
45.
go back to reference Curtis AR, Fey C, Morris CM. Mutation in the gene encoding ferritin light polypeptide causes dominant adult-onset basal ganglia disease. Nat Genet. 2001;28(4):350–4.CrossRefPubMed Curtis AR, Fey C, Morris CM. Mutation in the gene encoding ferritin light polypeptide causes dominant adult-onset basal ganglia disease. Nat Genet. 2001;28(4):350–4.CrossRefPubMed
46.
go back to reference Bradbury MW. Transport of iron in the blood–brain-cerebrospinal fluid system. Neurochem. 1997;69(2):443–54.CrossRef Bradbury MW. Transport of iron in the blood–brain-cerebrospinal fluid system. Neurochem. 1997;69(2):443–54.CrossRef
47.
go back to reference Attieh ZK, Mukhopadhyay CK, Seshadri V. Ceruloplasmin ferroxidase activity stimulates cellular iron uptake by a trivalent cation-specific transport mechanism. J Biol Chem. 1999;274(2):1116–23.CrossRefPubMed Attieh ZK, Mukhopadhyay CK, Seshadri V. Ceruloplasmin ferroxidase activity stimulates cellular iron uptake by a trivalent cation-specific transport mechanism. J Biol Chem. 1999;274(2):1116–23.CrossRefPubMed
48.
go back to reference Hulet SW, Powers S, Connor JR. Distribution of transferrin and ferritin binding in normal and multiple sclerotic human brains. J Neurol Sci. 1999;165(1):48–55.CrossRefPubMed Hulet SW, Powers S, Connor JR. Distribution of transferrin and ferritin binding in normal and multiple sclerotic human brains. J Neurol Sci. 1999;165(1):48–55.CrossRefPubMed
49.
go back to reference Hallgren B. The effect of age on the nonhaemin iron in the human brain. JNeurochem. 1958;3:41–51.CrossRef Hallgren B. The effect of age on the nonhaemin iron in the human brain. JNeurochem. 1958;3:41–51.CrossRef
50.
go back to reference Connor JR, Snyder BS, Arosio P. A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer’s diseased brains. J Neurochem. 1995;65(2):717–24.CrossRefPubMed Connor JR, Snyder BS, Arosio P. A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer’s diseased brains. J Neurochem. 1995;65(2):717–24.CrossRefPubMed
51.
go back to reference Harrison PM, Arosio P. The ferritins: molecular properties, iron storage function and cellular regulation. Biochim Biophys Acta. 1996;1275(3):161–203.CrossRefPubMed Harrison PM, Arosio P. The ferritins: molecular properties, iron storage function and cellular regulation. Biochim Biophys Acta. 1996;1275(3):161–203.CrossRefPubMed
52.
go back to reference Levi S, Yewdall SJ, Harrison PM, et al. Evidence that H- and L-chains have cooperative roles in the iron-uptake mechanism of human ferritin. Bioehem J. 1992;288:591–6. Levi S, Yewdall SJ, Harrison PM, et al. Evidence that H- and L-chains have cooperative roles in the iron-uptake mechanism of human ferritin. Bioehem J. 1992;288:591–6.
53.
go back to reference Moos T, Morgan EH. Evidence for low molecular weight, non-transferrin-bound iron in rat brain and cerebrospinal fluid. J Neurosci Res. 1998;54(4):486–94.CrossRefPubMed Moos T, Morgan EH. Evidence for low molecular weight, non-transferrin-bound iron in rat brain and cerebrospinal fluid. J Neurosci Res. 1998;54(4):486–94.CrossRefPubMed
55.
go back to reference Cappellini MD, Cohen A, Piga A, Bejaoui M. A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with beta-thalassemia. Blood. 2006;107(9):3455–62.CrossRefPubMed Cappellini MD, Cohen A, Piga A, Bejaoui M. A phase 3 study of deferasirox (ICL670), a once-daily oral iron chelator, in patients with beta-thalassemia. Blood. 2006;107(9):3455–62.CrossRefPubMed
56.
go back to reference Qian ZM, Shen X. Brain iron transport and neurodegeneration. Trend Mol Med. 2001;7(3):103–8.CrossRef Qian ZM, Shen X. Brain iron transport and neurodegeneration. Trend Mol Med. 2001;7(3):103–8.CrossRef
57.
go back to reference Connor JR, Fine RE. Development of transferrin-positive oligodendrocytes in the rat central nervous system. J Neurosci Res. 1987;17(1):51–9.CrossRefPubMed Connor JR, Fine RE. Development of transferrin-positive oligodendrocytes in the rat central nervous system. J Neurosci Res. 1987;17(1):51–9.CrossRefPubMed
58.
go back to reference Reddy MK, Labhasetwar V. Nanoparticle-mediated delivery of superoxide dismutase to the brain: an effective strategy to reduce ischemia-reperfusion injury. FASEB J. 2009;23(5):1384–95.CrossRefPubMed Reddy MK, Labhasetwar V. Nanoparticle-mediated delivery of superoxide dismutase to the brain: an effective strategy to reduce ischemia-reperfusion injury. FASEB J. 2009;23(5):1384–95.CrossRefPubMed
59.
go back to reference Kikuchi K, Kawahara K, Tancharoen S. The free radical scavenger edaravone rescues rats from cerebral infarction by attenuating the release of high-mobility group box-1 in neuronal cells. J Pharmacol Exp Ther. 2009;329(3):865–74.CrossRefPubMed Kikuchi K, Kawahara K, Tancharoen S. The free radical scavenger edaravone rescues rats from cerebral infarction by attenuating the release of high-mobility group box-1 in neuronal cells. J Pharmacol Exp Ther. 2009;329(3):865–74.CrossRefPubMed
60.
go back to reference Hayashi H, Yano M, Fujita Y. Compound overload of copper and iron in patients with Wilson’s disease. Med Mod Morphol. 2006;39(3):121.CrossRef Hayashi H, Yano M, Fujita Y. Compound overload of copper and iron in patients with Wilson’s disease. Med Mod Morphol. 2006;39(3):121.CrossRef
61.
go back to reference Schenck JF, Zimmemrman EA. High-field magnetic resonance imaging of brain iron: birth of a biomarker? NMR Biomed. 2004;17(7):433–45.CrossRefPubMed Schenck JF, Zimmemrman EA. High-field magnetic resonance imaging of brain iron: birth of a biomarker? NMR Biomed. 2004;17(7):433–45.CrossRefPubMed
62.
go back to reference Stankiewicz JM, Neema M, Ceccarelli A. Iron and multiple sclerosis. Neurobiol Aging. 2014;35(9):S51–8.CrossRefPubMed Stankiewicz JM, Neema M, Ceccarelli A. Iron and multiple sclerosis. Neurobiol Aging. 2014;35(9):S51–8.CrossRefPubMed
63.
go back to reference Turner MR, Kiernan MC, Leigh PN, et al. Biomarkers in amyotrophic lateral sclerosis. Lancet Neurol. 2009;8(1):94–109.CrossRefPubMed Turner MR, Kiernan MC, Leigh PN, et al. Biomarkers in amyotrophic lateral sclerosis. Lancet Neurol. 2009;8(1):94–109.CrossRefPubMed
64.
go back to reference Sian-Hulsmann J, Mandel S, Youdim MB, et al. The relevance of iron in the pathogenesis of Parkinson’s disease. Neurochem. 2011;118(6):939–57.CrossRef Sian-Hulsmann J, Mandel S, Youdim MB, et al. The relevance of iron in the pathogenesis of Parkinson’s disease. Neurochem. 2011;118(6):939–57.CrossRef
65.
go back to reference Grenier D, Huot MP, Mayrand D. Iron-chelating activity of tetracyclines and its impact on the susceptibility of Actinobacillus actinomycetemcomitans to these antibiotics. Antimicrob Agents Chemother. 2000;44(3):763–6.CrossRefPubMedPubMedCentral Grenier D, Huot MP, Mayrand D. Iron-chelating activity of tetracyclines and its impact on the susceptibility of Actinobacillus actinomycetemcomitans to these antibiotics. Antimicrob Agents Chemother. 2000;44(3):763–6.CrossRefPubMedPubMedCentral
Metadata
Title
Effect of oxidative stress induced by intracranial iron overload on central pain after spinal cord injury
Authors
Fan Xing Meng
Jing Ming Hou
Tian Sheng Sun
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Orthopaedic Surgery and Research / Issue 1/2017
Electronic ISSN: 1749-799X
DOI
https://doi.org/10.1186/s13018-017-0526-y

Other articles of this Issue 1/2017

Journal of Orthopaedic Surgery and Research 1/2017 Go to the issue