Skip to main content
Top
Published in:

Open Access 01-12-2017 | Review

Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases

Authors: Marcela Cruz-Haces, Jonathan Tang, Glen Acosta, Joseph Fernandez, Riyi Shi

Published in: Translational Neurodegeneration | Issue 1/2017

Login to get access

Abstract

Traumatic brain injury is among the most common causes of death and disability in youth and young adults. In addition to the acute risk of morbidity with moderate to severe injuries, traumatic brain injury is associated with a number of chronic neurological and neuropsychiatric sequelae including neurodegenerative diseases such as Alzheimer’s disease and Parkinson’s disease. However, despite the high incidence of traumatic brain injuries and the established clinical correlation with neurodegeneration, the causative factors linking these processes have not yet been fully elucidated. Apart from removal from activity, few, if any prophylactic treatments against post-traumatic brain injury neurodegeneration exist. Therefore, it is imperative to understand the pathophysiological mechanisms of traumatic brain injury and neurodegeneration in order to identify potential factors that initiate neurodegenerative processes. Oxidative stress, neuroinflammation, and glutamatergic excitotoxicity have previously been implicated in both secondary brain injury and neurodegeneration. In particular, reactive oxygen species appear to be key in mediating molecular insult in neuroinflammation and excitotoxicity. As such, it is likely that post injury oxidative stress is a key mechanism which links traumatic brain injury to increased risk of neurodegeneration. Consequently, reactive oxygen species and their subsequent byproducts may serve as novel fluid markers for identification and monitoring of cellular damage. Furthermore, these reactive species may further serve as a suitable therapeutic target to reduce the risk of post-injury neurodegeneration and provide long term quality of life improvements for those suffering from traumatic brain injury.
Literature
2.
go back to reference Maas AI, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–41.PubMedCrossRef Maas AI, Stocchetti N, Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–41.PubMedCrossRef
3.
go back to reference McIntosh TK, Smith DH, Meaney DF, Kotapka MJ, Gennarelli TA, Graham DI. Neuropathological sequelae of traumatic brain injury: relationship to neurochemical and biomechanical mechanisms. Lab Investig J Tech Methods Pathol. 1996;74:315–42. McIntosh TK, Smith DH, Meaney DF, Kotapka MJ, Gennarelli TA, Graham DI. Neuropathological sequelae of traumatic brain injury: relationship to neurochemical and biomechanical mechanisms. Lab Investig J Tech Methods Pathol. 1996;74:315–42.
4.
go back to reference Sosin DM, Sniezek JE, Thurman DJ. Incidence of mild and moderate brain injury in the United States, 1991. Brain Inj. 1996;10:47–54.PubMedCrossRef Sosin DM, Sniezek JE, Thurman DJ. Incidence of mild and moderate brain injury in the United States, 1991. Brain Inj. 1996;10:47–54.PubMedCrossRef
6.
go back to reference Bales JW, Kline AE, Wagner AK, Dixon CE. Targeting dopamine in acute traumatic brain injury. Open Drug Discov J. 2010;2:119–28.PubMedPubMedCentral Bales JW, Kline AE, Wagner AK, Dixon CE. Targeting dopamine in acute traumatic brain injury. Open Drug Discov J. 2010;2:119–28.PubMedPubMedCentral
7.
go back to reference Masel BE, DeWitt DS. Traumatic brain injury: a disease process. Not an Event J Neurotrauma. 2010;27:1529–40.PubMedCrossRef Masel BE, DeWitt DS. Traumatic brain injury: a disease process. Not an Event J Neurotrauma. 2010;27:1529–40.PubMedCrossRef
8.
go back to reference Rimel RW, Giordani B, Barth JT, Boll TJ, Jane JA. Disability caused by minor head injury. Neurosurgery. 1981;9:221–8.PubMed Rimel RW, Giordani B, Barth JT, Boll TJ, Jane JA. Disability caused by minor head injury. Neurosurgery. 1981;9:221–8.PubMed
9.
go back to reference Riggio S, Wong M. Neurobehavioral sequelae of traumatic brain injury. Mt. Sinai J. Med. J. Transl. Pers. Med. 2009;76:163–72.CrossRef Riggio S, Wong M. Neurobehavioral sequelae of traumatic brain injury. Mt. Sinai J. Med. J. Transl. Pers. Med. 2009;76:163–72.CrossRef
10.
go back to reference Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of traumatic brain injury: a brief overview. J Head Trauma Rehabil. 2006;21:375–8.PubMedCrossRef Langlois JA, Rutland-Brown W, Wald MM. The epidemiology and impact of traumatic brain injury: a brief overview. J Head Trauma Rehabil. 2006;21:375–8.PubMedCrossRef
11.
go back to reference Uryu K, Chen X-H, Martinez D, Browne KD, Johnson VE, Graham DI, et al. Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans. Exp Neurol. 2007;208:185–92.PubMedPubMedCentralCrossRef Uryu K, Chen X-H, Martinez D, Browne KD, Johnson VE, Graham DI, et al. Multiple proteins implicated in neurodegenerative diseases accumulate in axons after brain trauma in humans. Exp Neurol. 2007;208:185–92.PubMedPubMedCentralCrossRef
13.
go back to reference Goldman SM, Tanner CM, Oakes D, Bhudhikanok GS, Gupta A, Langston JW. Head injury and Parkinson’s disease risk in twins. Ann Neurol. 2006;60:65–72.PubMedCrossRef Goldman SM, Tanner CM, Oakes D, Bhudhikanok GS, Gupta A, Langston JW. Head injury and Parkinson’s disease risk in twins. Ann Neurol. 2006;60:65–72.PubMedCrossRef
15.
go back to reference Kampfl A, Posmantur RM, Zhao X, Schmutzhard E, Clifton GL, Hayes RL. Mechanisms of Calpain proteolysis following traumatic brain injury: implications for pathology and therapy: a review and update. J Neurotrauma. 1997;14:121–34.PubMedCrossRef Kampfl A, Posmantur RM, Zhao X, Schmutzhard E, Clifton GL, Hayes RL. Mechanisms of Calpain proteolysis following traumatic brain injury: implications for pathology and therapy: a review and update. J Neurotrauma. 1997;14:121–34.PubMedCrossRef
16.
go back to reference Johnson VE, Stewart W, Smith DH. Widespread tau and Amyloid-Beta pathology many years after a single traumatic brain injury in humans: long-term AD-like pathology after single TBI. Brain Pathol. 2012;22:142–9.PubMedCrossRef Johnson VE, Stewart W, Smith DH. Widespread tau and Amyloid-Beta pathology many years after a single traumatic brain injury in humans: long-term AD-like pathology after single TBI. Brain Pathol. 2012;22:142–9.PubMedCrossRef
17.
go back to reference DeKosky ST, Ikonomovic MD, Gandy S. Traumatic brain injury — football, warfare, and long-term effects. N Engl J Med. 2010;363:1293–6.PubMedCrossRef DeKosky ST, Ikonomovic MD, Gandy S. Traumatic brain injury — football, warfare, and long-term effects. N Engl J Med. 2010;363:1293–6.PubMedCrossRef
18.
go back to reference Marion DW, Penrod LE, Kelsey SF, Obrist WD, Kochanek PM, Palmer AM, et al. Treatment of traumatic brain injury with moderate hypothermia. N Engl J Med. 1997;336:540–6.PubMedCrossRef Marion DW, Penrod LE, Kelsey SF, Obrist WD, Kochanek PM, Palmer AM, et al. Treatment of traumatic brain injury with moderate hypothermia. N Engl J Med. 1997;336:540–6.PubMedCrossRef
19.
go back to reference Faden AL, Demediuk P, Panter SS, Vink R. The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science. 1989;244:798.PubMedCrossRef Faden AL, Demediuk P, Panter SS, Vink R. The role of excitatory amino acids and NMDA receptors in traumatic brain injury. Science. 1989;244:798.PubMedCrossRef
20.
go back to reference Khanna S, Davis D, Peterson B, Fisher B, Tung H, O’Quigley J, et al. Use of hypertonic saline in the treatment of severe refractory posttraumatic intracranial hypertension in pediatric traumatic brain injury: Crit. Care Med. 2000;28:1144–51.CrossRef Khanna S, Davis D, Peterson B, Fisher B, Tung H, O’Quigley J, et al. Use of hypertonic saline in the treatment of severe refractory posttraumatic intracranial hypertension in pediatric traumatic brain injury: Crit. Care Med. 2000;28:1144–51.CrossRef
21.
go back to reference Hoge CW, McGurk D, Thomas JL, Cox AL, Engel CC, Castro CA. Mild traumatic brain injury in U.S. soldiers returning from Iraq. N Engl J Med. 2008;358:453–63.PubMedCrossRef Hoge CW, McGurk D, Thomas JL, Cox AL, Engel CC, Castro CA. Mild traumatic brain injury in U.S. soldiers returning from Iraq. N Engl J Med. 2008;358:453–63.PubMedCrossRef
22.
go back to reference Rutland-Brown W, Langlois JA, Thomas KE, Xi YL, et al. Incidence of traumatic brain injury in the United States, 2003. J Head Trauma Rehabil. 2006;21:544.PubMedCrossRef Rutland-Brown W, Langlois JA, Thomas KE, Xi YL, et al. Incidence of traumatic brain injury in the United States, 2003. J Head Trauma Rehabil. 2006;21:544.PubMedCrossRef
23.
go back to reference Fu TS, Jing R, McFaull SR, Cusimano MD. Health & Economic Burden of traumatic brain injury in the emergency Department. Can J Neurol Sci. 2016;43:238–47.PubMedCrossRef Fu TS, Jing R, McFaull SR, Cusimano MD. Health & Economic Burden of traumatic brain injury in the emergency Department. Can J Neurol Sci. 2016;43:238–47.PubMedCrossRef
24.
go back to reference Jordan BD. The clinical spectrum of sport-related traumatic brain injury. Nat Rev Neurol. 2013;9:222–30.PubMedCrossRef Jordan BD. The clinical spectrum of sport-related traumatic brain injury. Nat Rev Neurol. 2013;9:222–30.PubMedCrossRef
25.
go back to reference Cernak I, Noble-Haeusslein LJ. Traumatic brain injury: an overview of pathobiology with emphasis on military populations. J Cereb Blood Flow Metab. 2010;30:255–66.PubMedCrossRef Cernak I, Noble-Haeusslein LJ. Traumatic brain injury: an overview of pathobiology with emphasis on military populations. J Cereb Blood Flow Metab. 2010;30:255–66.PubMedCrossRef
26.
go back to reference Malec JF, Brown AW, Leibson CL, Flaada JT, Mandrekar JN, Diehl NN, et al. The Mayo classification system for traumatic brain injury severity. J Neurotrauma. 2007;24:1417–24.PubMedCrossRef Malec JF, Brown AW, Leibson CL, Flaada JT, Mandrekar JN, Diehl NN, et al. The Mayo classification system for traumatic brain injury severity. J Neurotrauma. 2007;24:1417–24.PubMedCrossRef
27.
go back to reference Graham DI, Adams JH, Nicoll JAR, Maxwell WL, Gennarelli TA. The nature, distribution and causes of traumatic brain injury. Brain Pathol. 1995;5:397–406.PubMedCrossRef Graham DI, Adams JH, Nicoll JAR, Maxwell WL, Gennarelli TA. The nature, distribution and causes of traumatic brain injury. Brain Pathol. 1995;5:397–406.PubMedCrossRef
28.
29.
go back to reference Carroll L, Cassidy JD, Peloso P, Borg J, Von Holst H, Holm L, et al. Prognosis for mild traumatic brain injury: results of the WHO collaborating Centre task force on mild traumatic brain injury. J Rehabil Med. 2004;36:84–105.CrossRef Carroll L, Cassidy JD, Peloso P, Borg J, Von Holst H, Holm L, et al. Prognosis for mild traumatic brain injury: results of the WHO collaborating Centre task force on mild traumatic brain injury. J Rehabil Med. 2004;36:84–105.CrossRef
30.
go back to reference Summers CR, Ivins B, Schwab KA. Traumatic brain injury in the United States: an epidemiologic overview. Mt. Sinai J. Med. J. Transl. Pers. Med. 2009;76:105–10.CrossRef Summers CR, Ivins B, Schwab KA. Traumatic brain injury in the United States: an epidemiologic overview. Mt. Sinai J. Med. J. Transl. Pers. Med. 2009;76:105–10.CrossRef
31.
go back to reference Stern RA, Riley DO, Daneshvar DH, Nowinski CJ, Cantu RC, McKee AC. Long-term consequences of repetitive brain trauma: chronic traumatic encephalopathy. PM&R. 2011;3:S460–7.CrossRef Stern RA, Riley DO, Daneshvar DH, Nowinski CJ, Cantu RC, McKee AC. Long-term consequences of repetitive brain trauma: chronic traumatic encephalopathy. PM&R. 2011;3:S460–7.CrossRef
32.
go back to reference Cernak I, Wang Z, Jiang J, Bian X, Savic J. Ultrastructural and functional characteristics of blast injury-induced neurotrauma. J Trauma Acute Care Surg. 2001;50:695–706.CrossRef Cernak I, Wang Z, Jiang J, Bian X, Savic J. Ultrastructural and functional characteristics of blast injury-induced neurotrauma. J Trauma Acute Care Surg. 2001;50:695–706.CrossRef
33.
go back to reference Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg. 1991;75:15–26.PubMedCrossRef Tator CH, Fehlings MG. Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. J Neurosurg. 1991;75:15–26.PubMedCrossRef
35.
go back to reference Ommaya AK, Grubb RL Jr, Naumann RA. Coup and contre-coup injury: observations on the mechanics of visible brain injuries in the rhesus monkey. J Neurosurg. 1971;35:503–16.PubMedCrossRef Ommaya AK, Grubb RL Jr, Naumann RA. Coup and contre-coup injury: observations on the mechanics of visible brain injuries in the rhesus monkey. J Neurosurg. 1971;35:503–16.PubMedCrossRef
36.
go back to reference Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA, Hartl R, et al. Guidelines for the management of severe traumatic brain injury. VI. Indications for intracranial pressure monitoring. J Neurotrauma. 2007;24(Suppl 1):S37–44.PubMed Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA, Hartl R, et al. Guidelines for the management of severe traumatic brain injury. VI. Indications for intracranial pressure monitoring. J Neurotrauma. 2007;24(Suppl 1):S37–44.PubMed
37.
go back to reference Greve MW, Zink BJ. Pathophysiology of traumatic brain injury. Mt Sinai J Med J Transl Pers Med. 2009;76:97–104.CrossRef Greve MW, Zink BJ. Pathophysiology of traumatic brain injury. Mt Sinai J Med J Transl Pers Med. 2009;76:97–104.CrossRef
38.
go back to reference Harris JL, Yeh H-W, Choi I-Y, Lee P, Berman NE, Swerdlow RH, et al. Altered neurochemical profile after traumatic brain injury: 1H-MRS biomarkers of pathological mechanisms. J Cereb Blood Flow Metab. 2012;32:2122–34.PubMedPubMedCentralCrossRef Harris JL, Yeh H-W, Choi I-Y, Lee P, Berman NE, Swerdlow RH, et al. Altered neurochemical profile after traumatic brain injury: 1H-MRS biomarkers of pathological mechanisms. J Cereb Blood Flow Metab. 2012;32:2122–34.PubMedPubMedCentralCrossRef
39.
go back to reference Bang SA, Song YS, Moon BS, Lee BC, Lee H, Kim J-M, et al. Neuropsychological, metabolic, and GABA A receptor studies in subjects with repetitive traumatic brain injury. J Neurotrauma. 2016;33:1005–14.PubMedCrossRef Bang SA, Song YS, Moon BS, Lee BC, Lee H, Kim J-M, et al. Neuropsychological, metabolic, and GABA A receptor studies in subjects with repetitive traumatic brain injury. J Neurotrauma. 2016;33:1005–14.PubMedCrossRef
40.
go back to reference Bose R, Schnell CL, Pinsky C, Zitko V. Effects of excitotoxins on free radical indices in mouse brain. Toxicol Lett. 1992;60:211–9.PubMedCrossRef Bose R, Schnell CL, Pinsky C, Zitko V. Effects of excitotoxins on free radical indices in mouse brain. Toxicol Lett. 1992;60:211–9.PubMedCrossRef
41.
go back to reference Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, et al. Traumatic brain injury: oxidative stress and Neuroprotection. Antioxid Redox Signal. 2013;19:836–53.PubMedCrossRef Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, et al. Traumatic brain injury: oxidative stress and Neuroprotection. Antioxid Redox Signal. 2013;19:836–53.PubMedCrossRef
42.
43.
go back to reference Globus MY-T, Alonso O, Dietrich WD, Busto R, Ginsberg MD. Glutamate release and free radical production following brain injury: effects of posttraumatic hypothermia. J Neurochem. 2002;65:1704–11.CrossRef Globus MY-T, Alonso O, Dietrich WD, Busto R, Ginsberg MD. Glutamate release and free radical production following brain injury: effects of posttraumatic hypothermia. J Neurochem. 2002;65:1704–11.CrossRef
44.
go back to reference Leist M, Volbracht C, Kühnle S, Fava E, Ferrando-May E, Nicotera P. Caspase-mediated apoptosis in neuronal excitotoxicity triggered by nitric oxide. Mol Med. 1997;3:750–64.PubMedPubMedCentral Leist M, Volbracht C, Kühnle S, Fava E, Ferrando-May E, Nicotera P. Caspase-mediated apoptosis in neuronal excitotoxicity triggered by nitric oxide. Mol Med. 1997;3:750–64.PubMedPubMedCentral
45.
go back to reference Zhang Y, Bhavnani BR. Glutamate-induced apoptosis in neuronal cells is mediated via caspase-dependent and independent mechanisms involving calpain and caspase-3 proteases as well as apoptosis inducing factor (AIF) and this process is inhibited by equine estrogens. BMC Neurosci. 2006;7:49.PubMedPubMedCentralCrossRef Zhang Y, Bhavnani BR. Glutamate-induced apoptosis in neuronal cells is mediated via caspase-dependent and independent mechanisms involving calpain and caspase-3 proteases as well as apoptosis inducing factor (AIF) and this process is inhibited by equine estrogens. BMC Neurosci. 2006;7:49.PubMedPubMedCentralCrossRef
46.
go back to reference DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. J Neurochem. 139:136–53. DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. J Neurochem. 139:136–53.
47.
go back to reference Bayir H, Marion DW, Puccio AM, Wisniewski SR, Janesko KL, Clark RSB, et al. Marked gender effect on lipid peroxidation after severe traumatic brain injury in adult patients. J Neurotrauma. 2004;21:1–8.PubMedCrossRef Bayir H, Marion DW, Puccio AM, Wisniewski SR, Janesko KL, Clark RSB, et al. Marked gender effect on lipid peroxidation after severe traumatic brain injury in adult patients. J Neurotrauma. 2004;21:1–8.PubMedCrossRef
48.
go back to reference Negre-Salvayre A, Coatrieux C, Ingueneau C, Salvayre R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol. 2008;153:6–20.PubMedCrossRef Negre-Salvayre A, Coatrieux C, Ingueneau C, Salvayre R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol. 2008;153:6–20.PubMedCrossRef
49.
go back to reference Stevens JF, Maier CS. Acrolein: sources, metabolism, and biomolecular interactions relevant to human health and disease. Mol Nutr Food Res. 2008;52:7–25.PubMedPubMedCentralCrossRef Stevens JF, Maier CS. Acrolein: sources, metabolism, and biomolecular interactions relevant to human health and disease. Mol Nutr Food Res. 2008;52:7–25.PubMedPubMedCentralCrossRef
50.
go back to reference Ahmed FA, Kamnaksh A, Kovesdi E, Long JB, Agoston DV. Long-term consequences of single and multiple mild blast exposure on select physiological parameters and blood-based biomarkers. Electrophoresis. 2013;34:2229–33.PubMedCrossRef Ahmed FA, Kamnaksh A, Kovesdi E, Long JB, Agoston DV. Long-term consequences of single and multiple mild blast exposure on select physiological parameters and blood-based biomarkers. Electrophoresis. 2013;34:2229–33.PubMedCrossRef
51.
go back to reference Zheng L, Park J, Walls M, Tully M, Jannasch A, Cooper B, et al. Determination of urine 3-HPMA, a stable acrolein metabolite in a rat model of spinal cord injury. J Neurotrauma. 2013;30:1334–41.PubMedPubMedCentralCrossRef Zheng L, Park J, Walls M, Tully M, Jannasch A, Cooper B, et al. Determination of urine 3-HPMA, a stable acrolein metabolite in a rat model of spinal cord injury. J Neurotrauma. 2013;30:1334–41.PubMedPubMedCentralCrossRef
52.
go back to reference Uchida K, Kanematsu M, Sakai K, Matsuda T, Hattori N, Mizuno Y, et al. Protein-bound acrolein: potential markers for oxidative stress. Proc Natl Acad Sci U A. 1998;95:4882–7.CrossRef Uchida K, Kanematsu M, Sakai K, Matsuda T, Hattori N, Mizuno Y, et al. Protein-bound acrolein: potential markers for oxidative stress. Proc Natl Acad Sci U A. 1998;95:4882–7.CrossRef
53.
go back to reference Walls MK, Race N, Zheng L, Vega-Alvarez SM, Acosta G, Park J, et al. Structural and biochemical abnormalities in the absence of acute deficits in mild primary blast-induced head trauma. J Neurosurg. 2016;124:675–86.PubMedCrossRef Walls MK, Race N, Zheng L, Vega-Alvarez SM, Acosta G, Park J, et al. Structural and biochemical abnormalities in the absence of acute deficits in mild primary blast-induced head trauma. J Neurosurg. 2016;124:675–86.PubMedCrossRef
54.
go back to reference Mackay GM, Forrest CM, Stoy N, Christofides J, Egerton M, Stone TW, et al. Tryptophan metabolism and oxidative stress in patients with chronic brain injury. Eur J Neurol. 2006;13:30–42.PubMedCrossRef Mackay GM, Forrest CM, Stoy N, Christofides J, Egerton M, Stone TW, et al. Tryptophan metabolism and oxidative stress in patients with chronic brain injury. Eur J Neurol. 2006;13:30–42.PubMedCrossRef
55.
go back to reference Hoffer ME, Balaban C, Slade MD, Tsao JW, Hoffer B. Amelioration of acute Sequelae of blast induced mild traumatic brain injury by N-acetyl Cysteine: a double-blind, placebo controlled study. PLoS One. 2013;8 Hoffer ME, Balaban C, Slade MD, Tsao JW, Hoffer B. Amelioration of acute Sequelae of blast induced mild traumatic brain injury by N-acetyl Cysteine: a double-blind, placebo controlled study. PLoS One. 2013;8
56.
57.
go back to reference Kwiatkowski TJ, Bosco DA, LeClerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS/TLS Gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8.PubMedCrossRef Kwiatkowski TJ, Bosco DA, LeClerc AL, Tamrazian E, Vanderburg CR, Russ C, et al. Mutations in the FUS/TLS Gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8.PubMedCrossRef
58.
go back to reference Ajroud-Driss S, Siddique T. Sporadic and hereditary amyotrophic lateral sclerosis (ALS). Biochim. Biophys. Acta BBA-Mol. Basis Dis. 1852;2015:679–84. Ajroud-Driss S, Siddique T. Sporadic and hereditary amyotrophic lateral sclerosis (ALS). Biochim. Biophys. Acta BBA-Mol. Basis Dis. 1852;2015:679–84.
59.
60.
go back to reference Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, et al. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int. 2015;6:171.PubMedPubMedCentralCrossRef Zarei S, Carr K, Reiley L, Diaz K, Guerra O, Altamirano PF, et al. A comprehensive review of amyotrophic lateral sclerosis. Surg Neurol Int. 2015;6:171.PubMedPubMedCentralCrossRef
62.
go back to reference Forsberg K, Andersen PM, Marklund SL, Brannstrom T. Glial nuclear aggregates of superoxide dismutase-1 are regularly present in patients with amyotrophic lateral sclerosis. Acta Neuropathol. 2011;121:623–34.PubMedPubMedCentralCrossRef Forsberg K, Andersen PM, Marklund SL, Brannstrom T. Glial nuclear aggregates of superoxide dismutase-1 are regularly present in patients with amyotrophic lateral sclerosis. Acta Neuropathol. 2011;121:623–34.PubMedPubMedCentralCrossRef
64.
go back to reference Bruijn LI. Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science. 1998;281:1851–4.PubMedCrossRef Bruijn LI. Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science. 1998;281:1851–4.PubMedCrossRef
65.
66.
go back to reference Smith MA, Rottkamp CA, Nunomura A, Raina AK, Perry G. Oxidative stress in Alzheimer’s disease. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2000;1502:139–44.CrossRef Smith MA, Rottkamp CA, Nunomura A, Raina AK, Perry G. Oxidative stress in Alzheimer’s disease. Biochim. Biophys. Acta BBA-Mol. Basis Dis. 2000;1502:139–44.CrossRef
67.
go back to reference Barnham KJ, Masters CL, Bush AI. Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov. 2004;3:205–14.PubMedCrossRef Barnham KJ, Masters CL, Bush AI. Neurodegenerative diseases and oxidative stress. Nat Rev Drug Discov. 2004;3:205–14.PubMedCrossRef
68.
go back to reference Coyle J, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993;262:689–95.PubMedCrossRef Coyle J, Puttfarcken P. Oxidative stress, glutamate, and neurodegenerative disorders. Science. 1993;262:689–95.PubMedCrossRef
69.
go back to reference Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatanpaa K, et al. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc Natl Acad Sci. 2004;101:2070–5.PubMedPubMedCentralCrossRef Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatanpaa K, et al. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc Natl Acad Sci. 2004;101:2070–5.PubMedPubMedCentralCrossRef
70.
go back to reference Butterfield DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress1,2 1Guest editors: mark a. Smith and George Perry 2This article is part of a series of reviews on “causes and consequences of oxidative stress in Alzheimer’s disease.” the full list of papers may be found on the homepage of the journal. Free Radic Biol Med. 2002;32:1050–60.PubMedCrossRef Butterfield DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress1,2 1Guest editors: mark a. Smith and George Perry 2This article is part of a series of reviews on “causes and consequences of oxidative stress in Alzheimer’s disease.” the full list of papers may be found on the homepage of the journal. Free Radic Biol Med. 2002;32:1050–60.PubMedCrossRef
72.
go back to reference Ebadi M, Srinivasan SK, Baxi MD. Oxidative stress and antioxidant therapy in Parkinson’s disease. Prog Neurobiol. 1996;48:1–19.PubMedCrossRef Ebadi M, Srinivasan SK, Baxi MD. Oxidative stress and antioxidant therapy in Parkinson’s disease. Prog Neurobiol. 1996;48:1–19.PubMedCrossRef
73.
go back to reference Jenner P, Dexter DT, Sian J, Schapira AHV, Marsden CD. The Royal Kings and Queens Parkinson’s disease research group. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental lewy body disease. Ann. Neurol. 1992;32:S82–7. Jenner P, Dexter DT, Sian J, Schapira AHV, Marsden CD. The Royal Kings and Queens Parkinson’s disease research group. Oxidative stress as a cause of nigral cell death in Parkinson’s disease and incidental lewy body disease. Ann. Neurol. 1992;32:S82–7.
74.
go back to reference Jenner P, Olanow CW. Oxidative stress and the pathogenesis of Parkinson’s disease. Neurology. 1996;47:161S–70S.CrossRef Jenner P, Olanow CW. Oxidative stress and the pathogenesis of Parkinson’s disease. Neurology. 1996;47:161S–70S.CrossRef
76.
go back to reference Hald A, Lotharius J. Oxidative stress and inflammation in Parkinson’s disease: is there a causal link? Exp Neurol. 2005;193:279–90.PubMedCrossRef Hald A, Lotharius J. Oxidative stress and inflammation in Parkinson’s disease: is there a causal link? Exp Neurol. 2005;193:279–90.PubMedCrossRef
77.
go back to reference Bizzozero OA. Protein carbonylation in neurodegenerative and demyelinating CNS diseases. Handb. Neurochem. Mol. Neurobiol. Brain spinal cord. Trauma. 2009:543–62. Bizzozero OA. Protein carbonylation in neurodegenerative and demyelinating CNS diseases. Handb. Neurochem. Mol. Neurobiol. Brain spinal cord. Trauma. 2009:543–62.
78.
go back to reference Kabashi E, Valdmanis PN, Dion P, Rouleau GA. Oxidized/misfolded superoxide dismutase-1: the cause of all amyotrophic lateral sclerosis? Ann Neurol. 2007;62:553–9.PubMedCrossRef Kabashi E, Valdmanis PN, Dion P, Rouleau GA. Oxidized/misfolded superoxide dismutase-1: the cause of all amyotrophic lateral sclerosis? Ann Neurol. 2007;62:553–9.PubMedCrossRef
79.
go back to reference Barber SC, Mead RJ, Shaw PJ. Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim Biophys Acta BBA-Mol Basis Dis. 2006;1762:1051–67.CrossRef Barber SC, Mead RJ, Shaw PJ. Oxidative stress in ALS: a mechanism of neurodegeneration and a therapeutic target. Biochim Biophys Acta BBA-Mol Basis Dis. 2006;1762:1051–67.CrossRef
81.
go back to reference Beal MF. Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol. 1995;38:357–66.PubMedCrossRef Beal MF. Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol. 1995;38:357–66.PubMedCrossRef
82.
go back to reference Editorial KR. The prevalence and malignancy of Alzheimer disease. A major killer. Arch. Neurol. 1976;33:217–8. Editorial KR. The prevalence and malignancy of Alzheimer disease. A major killer. Arch. Neurol. 1976;33:217–8.
84.
go back to reference Wang H-Y. Beta -Amyloid1-42 binds to alpha 7 nicotinic acetylcholine receptor with high affinity. IMPLICATIONS FOR ALZHEIMER’S DISEASE PATHOLOGY. J Biol Chem. 2000;275:5626–32.PubMedCrossRef Wang H-Y. Beta -Amyloid1-42 binds to alpha 7 nicotinic acetylcholine receptor with high affinity. IMPLICATIONS FOR ALZHEIMER’S DISEASE PATHOLOGY. J Biol Chem. 2000;275:5626–32.PubMedCrossRef
85.
go back to reference Hardy J. The Amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6.PubMedCrossRef Hardy J. The Amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6.PubMedCrossRef
86.
go back to reference Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med. 1997;23:134–47.PubMedCrossRef Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med. 1997;23:134–47.PubMedCrossRef
87.
go back to reference Williams TI, Lynn BC, Markesbery WR, Lovell MA. Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer’s disease. Neurobiol Aging. 2006;27:1094–9.PubMedCrossRef Williams TI, Lynn BC, Markesbery WR, Lovell MA. Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimer’s disease. Neurobiol Aging. 2006;27:1094–9.PubMedCrossRef
88.
go back to reference Smith CD, Carney JM, Starke-Reed PE, Oliver CN, Stadtman ER, Floyd RA, et al. Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci. 1991;88:10540–3.PubMedPubMedCentralCrossRef Smith CD, Carney JM, Starke-Reed PE, Oliver CN, Stadtman ER, Floyd RA, et al. Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci. 1991;88:10540–3.PubMedPubMedCentralCrossRef
89.
go back to reference Gabbita SP, Lovell MA, Markesbery WR. Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem. 2002;71:2034–40.CrossRef Gabbita SP, Lovell MA, Markesbery WR. Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem. 2002;71:2034–40.CrossRef
90.
go back to reference Zemlan FP, Thienhaus OJ, Bosmann HB. Superoxide dismutase activity in Alzheimer’s disease: possible mechanism for paired helical filament formation. Brain Res. 1989;476:160–2.PubMedCrossRef Zemlan FP, Thienhaus OJ, Bosmann HB. Superoxide dismutase activity in Alzheimer’s disease: possible mechanism for paired helical filament formation. Brain Res. 1989;476:160–2.PubMedCrossRef
91.
go back to reference Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF, et al. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease. Proc Natl Acad Sci. 1994;91:3270–4.PubMedPubMedCentralCrossRef Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF, et al. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease. Proc Natl Acad Sci. 1994;91:3270–4.PubMedPubMedCentralCrossRef
92.
go back to reference Varadarajan S, Yatin S, Aksenova M, Butterfield DA. Review: Alzheimer’s Amyloid β-peptide-associated free radical oxidative stress and neurotoxicity. J Struct Biol. 2000;130:184–208.PubMedCrossRef Varadarajan S, Yatin S, Aksenova M, Butterfield DA. Review: Alzheimer’s Amyloid β-peptide-associated free radical oxidative stress and neurotoxicity. J Struct Biol. 2000;130:184–208.PubMedCrossRef
93.
go back to reference Allan BD. Amyloid β-peptide (1-42)-induced oxidative stress and neurotoxicity: implications for Neurodegeneration in Alzheimer’s disease brain. A Review Free Radic Res. 2002;36:1307–13.CrossRef Allan BD. Amyloid β-peptide (1-42)-induced oxidative stress and neurotoxicity: implications for Neurodegeneration in Alzheimer’s disease brain. A Review Free Radic Res. 2002;36:1307–13.CrossRef
94.
go back to reference Ramirez BG. Prevention of Alzheimer’s disease pathology by Cannabinoids: Neuroprotection mediated by blockade of Microglial activation. J Neurosci. 2005;25:1904–13.PubMedCrossRef Ramirez BG. Prevention of Alzheimer’s disease pathology by Cannabinoids: Neuroprotection mediated by blockade of Microglial activation. J Neurosci. 2005;25:1904–13.PubMedCrossRef
96.
go back to reference Bona D, Scapagnini G, Candore G, Castiglia L, Colonna-Romano G, Duro G, et al. Immune-inflammatory responses and oxidative stress in Alzheimers disease: therapeutic implications. Curr Pharm Des. 2010;16:684–91.PubMedCrossRef Bona D, Scapagnini G, Candore G, Castiglia L, Colonna-Romano G, Duro G, et al. Immune-inflammatory responses and oxidative stress in Alzheimers disease: therapeutic implications. Curr Pharm Des. 2010;16:684–91.PubMedCrossRef
97.
go back to reference Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJM, Rozemuller JM, Veerhuis R, et al. Neuroinflammation in Alzheimer’s disease and prion disease. Glia. 2002;40:232–9.PubMedCrossRef Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJM, Rozemuller JM, Veerhuis R, et al. Neuroinflammation in Alzheimer’s disease and prion disease. Glia. 2002;40:232–9.PubMedCrossRef
98.
go back to reference Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.PubMedCrossRef Heneka MT, Carson MJ, Khoury JE, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.PubMedCrossRef
99.
go back to reference Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective-Amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci. 2008;28:8354–60.PubMedPubMedCentralCrossRef Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective-Amyloid clearance pathways in aging Alzheimer’s disease mice. J Neurosci. 2008;28:8354–60.PubMedPubMedCentralCrossRef
100.
go back to reference Hynd M. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer?S disease. Neurochem Int. 2004;45:583–95.PubMedCrossRef Hynd M. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer?S disease. Neurochem Int. 2004;45:583–95.PubMedCrossRef
101.
go back to reference DeLeo JA, Yezierski RP. The role of neuroinflammation and neuroimmune activation in persistent pain. Pain. 2001;90:1–6.PubMedCrossRef DeLeo JA, Yezierski RP. The role of neuroinflammation and neuroimmune activation in persistent pain. Pain. 2001;90:1–6.PubMedCrossRef
102.
go back to reference Stern D. Non-enzymatically glycated tau in Alzheimer’s disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid b-peptide. Nat Med. 1995;1:693–9.CrossRef Stern D. Non-enzymatically glycated tau in Alzheimer’s disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid b-peptide. Nat Med. 1995;1:693–9.CrossRef
103.
go back to reference Lauderback CM, Hackett JM, Huang FF, Keller JN, Szweda LI, Markesbery WR, et al. The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer’s disease brain: the role of Aβ1-42: Aβ1-42 and HNE binding to GLT-1 in AD brain. J Neurochem. 2001;78:413–6.PubMedCrossRef Lauderback CM, Hackett JM, Huang FF, Keller JN, Szweda LI, Markesbery WR, et al. The glial glutamate transporter, GLT-1, is oxidatively modified by 4-hydroxy-2-nonenal in the Alzheimer’s disease brain: the role of Aβ1-42: Aβ1-42 and HNE binding to GLT-1 in AD brain. J Neurochem. 2001;78:413–6.PubMedCrossRef
105.
go back to reference McKee AC, Stein TD, Nowinski CJ, Stern RA, Daneshvar DH, Alvarez VE, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 2013;136:43–64.PubMedCrossRef McKee AC, Stein TD, Nowinski CJ, Stern RA, Daneshvar DH, Alvarez VE, et al. The spectrum of disease in chronic traumatic encephalopathy. Brain. 2013;136:43–64.PubMedCrossRef
106.
go back to reference Blaylock RL, Maroon J, et al. Immunoexcitotoxicity as a central mechanism in chronic traumatic encephalopathy-a unifying hypothesis. Surg Neurol Int. 2011;2:4103. Blaylock RL, Maroon J, et al. Immunoexcitotoxicity as a central mechanism in chronic traumatic encephalopathy-a unifying hypothesis. Surg Neurol Int. 2011;2:4103.
107.
go back to reference McKee AC, Cantu RC, Nowinski CJ, Hedley-Whyte ET, Gavett BE, Budson AE, et al. Chronic traumatic encephalopathy in athletes: progressive Tauopathy after repetitive head injury. J Neuropathol Exp Neurol. 2009;68:709–35.PubMedPubMedCentralCrossRef McKee AC, Cantu RC, Nowinski CJ, Hedley-Whyte ET, Gavett BE, Budson AE, et al. Chronic traumatic encephalopathy in athletes: progressive Tauopathy after repetitive head injury. J Neuropathol Exp Neurol. 2009;68:709–35.PubMedPubMedCentralCrossRef
108.
go back to reference McKee AC, Gavett BE, Stern RA, Nowinski CJ, Cantu RC, Kowall NW, et al. TDP-43 Proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 2010;69:918–29.PubMedPubMedCentralCrossRef McKee AC, Gavett BE, Stern RA, Nowinski CJ, Cantu RC, Kowall NW, et al. TDP-43 Proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 2010;69:918–29.PubMedPubMedCentralCrossRef
109.
go back to reference Yi J, Padalino DJ, Chin LS, Montenegro P, Cantu RC. Chronic traumatic encephalopathy: Curr. Sports Med Rep. 2013;12:28–32.CrossRef Yi J, Padalino DJ, Chin LS, Montenegro P, Cantu RC. Chronic traumatic encephalopathy: Curr. Sports Med Rep. 2013;12:28–32.CrossRef
110.
go back to reference Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow E-M. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;156:1051–63.PubMedPubMedCentralCrossRef Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow E-M. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;156:1051–63.PubMedPubMedCentralCrossRef
111.
go back to reference Hebron ML, Lonskaya I, Sharpe K, Weerasinghe PPK, Algarzae NK, Shekoyan AR, et al. Parkin Ubiquitinates tar-DNA binding protein-43 (TDP-43) and promotes its Cytosolic accumulation via interaction with Histone Deacetylase 6 (HDAC6). J Biol Chem. 2013;288:4103–15.PubMedCrossRef Hebron ML, Lonskaya I, Sharpe K, Weerasinghe PPK, Algarzae NK, Shekoyan AR, et al. Parkin Ubiquitinates tar-DNA binding protein-43 (TDP-43) and promotes its Cytosolic accumulation via interaction with Histone Deacetylase 6 (HDAC6). J Biol Chem. 2013;288:4103–15.PubMedCrossRef
112.
go back to reference Bandyopadhyay U, Cotney J, Nagy M, Oh S, Leng J, Mahajan M, et al. RNA-Seq profiling of spinal cord motor neurons from a Presymptomatic SOD1 ALS mouse. PLoS One. 2013;8:e53575. Cai H, editorPubMedPubMedCentralCrossRef Bandyopadhyay U, Cotney J, Nagy M, Oh S, Leng J, Mahajan M, et al. RNA-Seq profiling of spinal cord motor neurons from a Presymptomatic SOD1 ALS mouse. PLoS One. 2013;8:e53575. Cai H, editorPubMedPubMedCentralCrossRef
113.
go back to reference Lucke-Wold BP, Turner RC, Logsdon AF, Bailes JE, Huber JD, Rosen CL. Linking traumatic brain injury to chronic traumatic encephalopathy: identification of potential mechanisms leading to Neurofibrillary tangle development. J Neurotrauma. 2014;31:1129–38.PubMedPubMedCentralCrossRef Lucke-Wold BP, Turner RC, Logsdon AF, Bailes JE, Huber JD, Rosen CL. Linking traumatic brain injury to chronic traumatic encephalopathy: identification of potential mechanisms leading to Neurofibrillary tangle development. J Neurotrauma. 2014;31:1129–38.PubMedPubMedCentralCrossRef
114.
go back to reference Saulle M, Greenwald BD. Chronic traumatic encephalopathy: a review. Rehabil Res Pract. 2012;2012:1–9. Saulle M, Greenwald BD. Chronic traumatic encephalopathy: a review. Rehabil Res Pract. 2012;2012:1–9.
115.
go back to reference Zhang Y, Dawson VL, Dawson TM. Oxidative stress and genetics in the pathogenesis of Parkinson’s disease. Neurobiol Dis. 2000;7:240–50.PubMedCrossRef Zhang Y, Dawson VL, Dawson TM. Oxidative stress and genetics in the pathogenesis of Parkinson’s disease. Neurobiol Dis. 2000;7:240–50.PubMedCrossRef
116.
go back to reference Atsmon-Raz Y, Miller Y. Non-Amyloid-beta component of human alpha-Synuclein Oligomers induces formation of new Abeta Oligomers: insight into the mechanisms that link Parkinson’s and Alzheimer’s diseases. ACS Chem Neurosci. 2016;7:46–55.PubMedCrossRef Atsmon-Raz Y, Miller Y. Non-Amyloid-beta component of human alpha-Synuclein Oligomers induces formation of new Abeta Oligomers: insight into the mechanisms that link Parkinson’s and Alzheimer’s diseases. ACS Chem Neurosci. 2016;7:46–55.PubMedCrossRef
117.
go back to reference Plotegher N, Bubacco L. Lysines, Achilles’ heel in alpha-synuclein conversion to a deadly neuronal endotoxin. Ageing Res Rev. 2016;26:62–71.PubMedCrossRef Plotegher N, Bubacco L. Lysines, Achilles’ heel in alpha-synuclein conversion to a deadly neuronal endotoxin. Ageing Res Rev. 2016;26:62–71.PubMedCrossRef
118.
go back to reference Shamoto-Nagai M, Maruyama W, Hashizume Y, Yoshida M, Osawa T, Riederer P, et al. In parkinsonian substantia nigra, α-synuclein is modified by acrolein, a lipid-peroxidation product, and accumulates in the dopamine neurons with inhibition of proteasome activity. J Neural Transm. 2007;114:1559–67.PubMedCrossRef Shamoto-Nagai M, Maruyama W, Hashizume Y, Yoshida M, Osawa T, Riederer P, et al. In parkinsonian substantia nigra, α-synuclein is modified by acrolein, a lipid-peroxidation product, and accumulates in the dopamine neurons with inhibition of proteasome activity. J Neural Transm. 2007;114:1559–67.PubMedCrossRef
119.
go back to reference Qin Z, Hu D, Han S, Reaney SH, Di Monte DA, Fink AL. Effect of 4-Hydroxy-2-nonenal modification on -Synuclein aggregation. J Biol Chem. 2007;282:5862–70.PubMedCrossRef Qin Z, Hu D, Han S, Reaney SH, Di Monte DA, Fink AL. Effect of 4-Hydroxy-2-nonenal modification on -Synuclein aggregation. J Biol Chem. 2007;282:5862–70.PubMedCrossRef
120.
go back to reference Zhelev Z, Bakalova R, Aoki I, Lazarova D, Saga T. Imaging of superoxide generation in the Dopaminergic area of the brain in Parkinson’s disease, using Mito-TEMPO. ACS Chem Neurosci. 2013;4:1439–45.PubMedPubMedCentralCrossRef Zhelev Z, Bakalova R, Aoki I, Lazarova D, Saga T. Imaging of superoxide generation in the Dopaminergic area of the brain in Parkinson’s disease, using Mito-TEMPO. ACS Chem Neurosci. 2013;4:1439–45.PubMedPubMedCentralCrossRef
121.
go back to reference Parihar MS, Parihar A, Fujita M, Hashimoto M, Ghafourifar P. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci. 2008;65:1272–84.PubMedCrossRef Parihar MS, Parihar A, Fujita M, Hashimoto M, Ghafourifar P. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci. 2008;65:1272–84.PubMedCrossRef
122.
go back to reference McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:S3–7.PubMedCrossRef McGeer PL, McGeer EG. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat Disord. 2004;10:S3–7.PubMedCrossRef
123.
go back to reference McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–91.PubMedCrossRef McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38:1285–91.PubMedCrossRef
124.
go back to reference Yamada T, McGeer EG, Schelper RL, Wszolek ZK, McGeer PL, Pfeiffer RF, et al. Histological and biochemical pathology in a family with autosomal dominant parkinsonism and dementia. Neurol Psychiatry Brain Res. 1993;2 Yamada T, McGeer EG, Schelper RL, Wszolek ZK, McGeer PL, Pfeiffer RF, et al. Histological and biochemical pathology in a family with autosomal dominant parkinsonism and dementia. Neurol Psychiatry Brain Res. 1993;2
125.
go back to reference McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,4-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.PubMedCrossRef McGeer PL, Schwab C, Parent A, Doudet D. Presence of reactive microglia in monkey substantia nigra years after 1-methyl-4-phenyl-1,2,3,4-tetrahydropyridine administration. Ann Neurol. 2003;54:599–604.PubMedCrossRef
126.
go back to reference Doble A. The pharmacology and mechanism of action of riluzole. Neurology. 1996;47:233S–41S.CrossRef Doble A. The pharmacology and mechanism of action of riluzole. Neurology. 1996;47:233S–41S.CrossRef
127.
go back to reference Oster S, Radad K, Scheller D, Hesse M, Balanzew W, Reichmann H, et al. Rotigotine protects against glutamate toxicity in primary dopaminergic cell culture. Eur J Pharmacol. 2014;724:31–42.PubMedCrossRef Oster S, Radad K, Scheller D, Hesse M, Balanzew W, Reichmann H, et al. Rotigotine protects against glutamate toxicity in primary dopaminergic cell culture. Eur J Pharmacol. 2014;724:31–42.PubMedCrossRef
128.
129.
go back to reference Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–72.PubMedCrossRef Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319:1668–72.PubMedCrossRef
130.
go back to reference Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med. 2002;33:337–49.PubMedCrossRef Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med. 2002;33:337–49.PubMedCrossRef
131.
go back to reference Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med. 2010;48:629–41.PubMedCrossRef Barber SC, Shaw PJ. Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. Free Radic Biol Med. 2010;48:629–41.PubMedCrossRef
132.
go back to reference Reaume AG, Elliott JL, Hoffman EK, Kowall NW, Ferrante RJ, Siwek DF, et al. Motor neurons in cu/Zn superoxide dismutase-deficient mice develop normally but exhibit enhanced cell death after axonal injury. Nat Genet. 1996;13:43–7.PubMedCrossRef Reaume AG, Elliott JL, Hoffman EK, Kowall NW, Ferrante RJ, Siwek DF, et al. Motor neurons in cu/Zn superoxide dismutase-deficient mice develop normally but exhibit enhanced cell death after axonal injury. Nat Genet. 1996;13:43–7.PubMedCrossRef
133.
go back to reference Liu J, Lillo C, Jonsson PA, Vande Velde C, Ward CM, Miller TM, et al. Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron. 2004;43:5–17.PubMedCrossRef Liu J, Lillo C, Jonsson PA, Vande Velde C, Ward CM, Miller TM, et al. Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron. 2004;43:5–17.PubMedCrossRef
134.
go back to reference Trotti D, Rolfs A, Danbolt NC, Brown RH, Hediger MA. SOD1 mutants linked to amyotrophic lateral sclerosis selectively inactivate a glial glutamate transporter. Nat Neurosci. 1999;2:427–33.PubMedCrossRef Trotti D, Rolfs A, Danbolt NC, Brown RH, Hediger MA. SOD1 mutants linked to amyotrophic lateral sclerosis selectively inactivate a glial glutamate transporter. Nat Neurosci. 1999;2:427–33.PubMedCrossRef
135.
136.
go back to reference Foran E, Trotti D. Glutamate transporters and the excitotoxic path to motor neuron degeneration in amyotrophic lateral sclerosis. Antioxid Redox Signal. 2009;11:1587–602.PubMedPubMedCentralCrossRef Foran E, Trotti D. Glutamate transporters and the excitotoxic path to motor neuron degeneration in amyotrophic lateral sclerosis. Antioxid Redox Signal. 2009;11:1587–602.PubMedPubMedCentralCrossRef
137.
go back to reference McGeer PL, McGeer EG. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002;26:459–70.PubMedCrossRef McGeer PL, McGeer EG. Inflammatory processes in amyotrophic lateral sclerosis. Muscle Nerve. 2002;26:459–70.PubMedCrossRef
138.
go back to reference Carbone M, Duty S, Rattray M. Riluzole elevates GLT-1 activity and levels in striatal astrocytes. Neurochem Int. 2012;60:31–8.PubMedCrossRef Carbone M, Duty S, Rattray M. Riluzole elevates GLT-1 activity and levels in striatal astrocytes. Neurochem Int. 2012;60:31–8.PubMedCrossRef
139.
go back to reference Wootz H, Hansson I, Korhonen L, Näpänkangas U, Lindholm D. Caspase-12 cleavage and increased oxidative stress during motoneuron degeneration in transgenic mouse model of ALS. Biochem Biophys Res Commun. 2004;322:281–6.PubMedCrossRef Wootz H, Hansson I, Korhonen L, Näpänkangas U, Lindholm D. Caspase-12 cleavage and increased oxidative stress during motoneuron degeneration in transgenic mouse model of ALS. Biochem Biophys Res Commun. 2004;322:281–6.PubMedCrossRef
140.
go back to reference Roberts GW, Gentleman SM, Lynch A, Graham DI. βA4 amyloid protein deposition in brain after head trauma. Lancet. 1991;338:1422–3.PubMedCrossRef Roberts GW, Gentleman SM, Lynch A, Graham DI. βA4 amyloid protein deposition in brain after head trauma. Lancet. 1991;338:1422–3.PubMedCrossRef
141.
go back to reference Lye TC, Shores EA. Traumatic brain injury as a risk factor for Alzheimer’s disease: a review. Neuropsychol Rev. 2000;10:115–29.PubMedCrossRef Lye TC, Shores EA. Traumatic brain injury as a risk factor for Alzheimer’s disease: a review. Neuropsychol Rev. 2000;10:115–29.PubMedCrossRef
142.
go back to reference Chiò A, Benzi G, Dossena M, Mutani R, Mora G. Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players. Brain. 2005;128:472.PubMedCrossRef Chiò A, Benzi G, Dossena M, Mutani R, Mora G. Severely increased risk of amyotrophic lateral sclerosis among Italian professional football players. Brain. 2005;128:472.PubMedCrossRef
143.
go back to reference Chio A, Calvo A, Dossena M, Ghiglione P, Mutani R, Mora G. ALS in Italian professional soccer players: the risk is still present and could be soccer-specific. Amyotroph Lateral Scler. 2009;10:205–9.PubMedCrossRef Chio A, Calvo A, Dossena M, Ghiglione P, Mutani R, Mora G. ALS in Italian professional soccer players: the risk is still present and could be soccer-specific. Amyotroph Lateral Scler. 2009;10:205–9.PubMedCrossRef
144.
go back to reference Armon C, Nelson LM. Is head trauma a risk factor for amyotrophic lateral sclerosis? An evidence based review. Amyotroph Lateral Scler. 2012;13:351–6.PubMedCrossRef Armon C, Nelson LM. Is head trauma a risk factor for amyotrophic lateral sclerosis? An evidence based review. Amyotroph Lateral Scler. 2012;13:351–6.PubMedCrossRef
145.
go back to reference Omalu BI, DeKosky ST, Minster RL, Kamboh MI, Hamilton RL, Wecht CH. Chronic traumatic encephalopathy in a National Football League Player. Neurosurgery. 2005;57:128–34.PubMedCrossRef Omalu BI, DeKosky ST, Minster RL, Kamboh MI, Hamilton RL, Wecht CH. Chronic traumatic encephalopathy in a National Football League Player. Neurosurgery. 2005;57:128–34.PubMedCrossRef
146.
go back to reference Smith DH, Uryu K, Saatman KE, Trojanowski JQ, McIntosh TK. Protein accumulation in traumatic brain injury. Neuro Molecular Med. 2003;4:59–72.CrossRef Smith DH, Uryu K, Saatman KE, Trojanowski JQ, McIntosh TK. Protein accumulation in traumatic brain injury. Neuro Molecular Med. 2003;4:59–72.CrossRef
147.
go back to reference Bramlett HM, Kraydieh S, Green EJ, Dietrich WD. Temporal and regional patterns of axonal damage following traumatic brain injury: a Beta-amyloid precursor protein Immunocytochemical study in rats. J Neuropathol Exp Neurol. 1997;56:1132–41.PubMedCrossRef Bramlett HM, Kraydieh S, Green EJ, Dietrich WD. Temporal and regional patterns of axonal damage following traumatic brain injury: a Beta-amyloid precursor protein Immunocytochemical study in rats. J Neuropathol Exp Neurol. 1997;56:1132–41.PubMedCrossRef
148.
go back to reference Goldman SM, Kamel F, Ross GW, Jewell SA, Bhudhikanok GS, Umbach D, et al. Head injury, alpha-synuclein Rep1, and Parkinson’s disease. Ann Neurol. 2012;71:40–8.PubMedPubMedCentralCrossRef Goldman SM, Kamel F, Ross GW, Jewell SA, Bhudhikanok GS, Umbach D, et al. Head injury, alpha-synuclein Rep1, and Parkinson’s disease. Ann Neurol. 2012;71:40–8.PubMedPubMedCentralCrossRef
149.
go back to reference Franz G, Beer R, Kampfl A, Engelhardt K, Schmutzhard E, Ulmer H, et al. Amyloid beta 1-42 and tau in cerebrospinal fluid after severe traumatic brain injury. Neurology. 2003;60:1457–61.PubMedCrossRef Franz G, Beer R, Kampfl A, Engelhardt K, Schmutzhard E, Ulmer H, et al. Amyloid beta 1-42 and tau in cerebrospinal fluid after severe traumatic brain injury. Neurology. 2003;60:1457–61.PubMedCrossRef
150.
go back to reference Tanzi R, Gusella J, Watkins P, Bruns G, St George-Hyslop P, Van Keuren M, et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science. 1987;235:880–4.PubMedCrossRef Tanzi R, Gusella J, Watkins P, Bruns G, St George-Hyslop P, Van Keuren M, et al. Amyloid beta protein gene: cDNA, mRNA distribution, and genetic linkage near the Alzheimer locus. Science. 1987;235:880–4.PubMedCrossRef
151.
go back to reference Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, Sandmann-Keil D, Rüb U. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson’s disease (preclinical and clinical stages). J Neurol. 2002;249:1–1.CrossRef Braak H, Del Tredici K, Bratzke H, Hamm-Clement J, Sandmann-Keil D, Rüb U. Staging of the intracerebral inclusion body pathology associated with idiopathic Parkinson’s disease (preclinical and clinical stages). J Neurol. 2002;249:1–1.CrossRef
152.
go back to reference Acosta SA, Tajiri N, de la Pena I, Bastawrous M, Sanberg PR, Kaneko Y, et al. Alpha-synuclein as a pathological link between chronic traumatic brain injury and Parkinson’s disease. J Cell Physiol. 2015;230:1024–32.PubMedPubMedCentralCrossRef Acosta SA, Tajiri N, de la Pena I, Bastawrous M, Sanberg PR, Kaneko Y, et al. Alpha-synuclein as a pathological link between chronic traumatic brain injury and Parkinson’s disease. J Cell Physiol. 2015;230:1024–32.PubMedPubMedCentralCrossRef
153.
go back to reference Gavett BE, Stern RA, McKee AC. Chronic traumatic encephalopathy: a potential late effect of sport-related concussive and Subconcussive head trauma. Clin Sports Med. 2011;30:179–88.PubMedPubMedCentralCrossRef Gavett BE, Stern RA, McKee AC. Chronic traumatic encephalopathy: a potential late effect of sport-related concussive and Subconcussive head trauma. Clin Sports Med. 2011;30:179–88.PubMedPubMedCentralCrossRef
154.
go back to reference Spillantini MG, Goedert M. Tau protein pathology in neurodegenerative diseases. Trends Neurosci. 1998;21:428–33.PubMedCrossRef Spillantini MG, Goedert M. Tau protein pathology in neurodegenerative diseases. Trends Neurosci. 1998;21:428–33.PubMedCrossRef
155.
go back to reference Pappolla MA, Chyan YJ, Omar TR, Hsiao K, Perry G, Smith MA, et al. Evidence of oxidative stress and in vivo neurotoxicity of beta-amyloid in a transgenic mouse model of Alzheimer’s disease: a chronic oxidative paradigm for testing antioxidant therapies in vivo. Am J Pathol. 1998;152:871.PubMedPubMedCentral Pappolla MA, Chyan YJ, Omar TR, Hsiao K, Perry G, Smith MA, et al. Evidence of oxidative stress and in vivo neurotoxicity of beta-amyloid in a transgenic mouse model of Alzheimer’s disease: a chronic oxidative paradigm for testing antioxidant therapies in vivo. Am J Pathol. 1998;152:871.PubMedPubMedCentral
156.
go back to reference Poppek D, Keck S, Ermak G, Jung T, Stolzing A, Ullrich O, et al. Phosphorylation inhibits turnover of the tau protein by the proteasome: influence of RCAN1 and oxidative stress. Biochem J. 2006;400:511–20.PubMedPubMedCentralCrossRef Poppek D, Keck S, Ermak G, Jung T, Stolzing A, Ullrich O, et al. Phosphorylation inhibits turnover of the tau protein by the proteasome: influence of RCAN1 and oxidative stress. Biochem J. 2006;400:511–20.PubMedPubMedCentralCrossRef
157.
go back to reference Moghe A, Ghare S, Lamoreau B, Mohammad M, Barve S, McClain C, et al. Molecular mechanisms of acrolein toxicity: relevance to human disease. Toxicol Sci. 2015;143:242–55.PubMedPubMedCentralCrossRef Moghe A, Ghare S, Lamoreau B, Mohammad M, Barve S, McClain C, et al. Molecular mechanisms of acrolein toxicity: relevance to human disease. Toxicol Sci. 2015;143:242–55.PubMedPubMedCentralCrossRef
158.
go back to reference Wood PL, Khan MA, Kulow SR, Mahmood SA, Moskal JR. Neurotoxicity of reactive aldehydes: the concept of “aldehyde load” as demonstrated by neuroprotection with hydroxylamines. Brain Res. 2006;1095:190–9.PubMedCrossRef Wood PL, Khan MA, Kulow SR, Mahmood SA, Moskal JR. Neurotoxicity of reactive aldehydes: the concept of “aldehyde load” as demonstrated by neuroprotection with hydroxylamines. Brain Res. 2006;1095:190–9.PubMedCrossRef
159.
160.
go back to reference Hicks RR, Smith DH, Lowenstein DH, Marie RS, McINTOSH TK. Mild experimental brain injury in the rat induces cognitive deficits associated with regional neuronal loss in the hippocampus. J Neurotrauma. 1993;10:405–14.PubMedCrossRef Hicks RR, Smith DH, Lowenstein DH, Marie RS, McINTOSH TK. Mild experimental brain injury in the rat induces cognitive deficits associated with regional neuronal loss in the hippocampus. J Neurotrauma. 1993;10:405–14.PubMedCrossRef
161.
go back to reference Šimić G, Lucassen PJ, Krsnik Ž, Krušlin B, Kostović I, Winblad B, et al. nNOS expression in reactive Astrocytes correlates with increased cell death related DNA damage in the hippocampus and Entorhinal cortex in Alzheimer’s disease. Exp Neurol. 2000;165:12–26.PubMedCrossRef Šimić G, Lucassen PJ, Krsnik Ž, Krušlin B, Kostović I, Winblad B, et al. nNOS expression in reactive Astrocytes correlates with increased cell death related DNA damage in the hippocampus and Entorhinal cortex in Alzheimer’s disease. Exp Neurol. 2000;165:12–26.PubMedCrossRef
162.
go back to reference Wilde EA, Bigler ED, Hunter JV, Fearing MA, Scheibel RS, Newsome MR, et al. Hippocampus, amygdala, and basal ganglia morphometrics in children after moderate-to-severe traumatic brain injury. Dev Med Child Neurol. 2007;49:294–9.PubMedCrossRef Wilde EA, Bigler ED, Hunter JV, Fearing MA, Scheibel RS, Newsome MR, et al. Hippocampus, amygdala, and basal ganglia morphometrics in children after moderate-to-severe traumatic brain injury. Dev Med Child Neurol. 2007;49:294–9.PubMedCrossRef
163.
go back to reference Carlsson M, Carlsson A. Interactions between glutamatergic and monoaminergic systems within the basal ganglia-implications for schizophrenia and Parkinson’s disease. Trends Neurosci. 1990;13:272–6.PubMedCrossRef Carlsson M, Carlsson A. Interactions between glutamatergic and monoaminergic systems within the basal ganglia-implications for schizophrenia and Parkinson’s disease. Trends Neurosci. 1990;13:272–6.PubMedCrossRef
164.
go back to reference Baugh CM, Stamm JM, Riley DO, Gavett BE, Shenton ME, Lin A, et al. Chronic traumatic encephalopathy: neurodegeneration following repetitive concussive and subconcussive brain trauma. Brain Imaging Behav. 2012;6:244–54.PubMedCrossRef Baugh CM, Stamm JM, Riley DO, Gavett BE, Shenton ME, Lin A, et al. Chronic traumatic encephalopathy: neurodegeneration following repetitive concussive and subconcussive brain trauma. Brain Imaging Behav. 2012;6:244–54.PubMedCrossRef
165.
go back to reference Brasure M, Lamberty GJ, Sayer NA, Nelson NW, MacDonald R, Ouellette J, et al. Multidisciplinary Postacute rehabilitation for moderate to severe traumatic brain injury in adults; 2012. p. ES1–ES20. Brasure M, Lamberty GJ, Sayer NA, Nelson NW, MacDonald R, Ouellette J, et al. Multidisciplinary Postacute rehabilitation for moderate to severe traumatic brain injury in adults; 2012. p. ES1–ES20.
166.
go back to reference Centers for Disease Control and Prevention (CDC). Traumatic brain injury in the United States: epidemiology and rehabilitation. 2015. Centers for Disease Control and Prevention (CDC). Traumatic brain injury in the United States: epidemiology and rehabilitation. 2015.
167.
go back to reference Menon DK, Schwab K, Wright DW, Maas AI, Demographics, Clinical Assessment Working Group of the I, et al. Position statement: definition of traumatic brain injury. Arch Phys Med Rehabil. 2010;91:1637–40.PubMedCrossRef Menon DK, Schwab K, Wright DW, Maas AI, Demographics, Clinical Assessment Working Group of the I, et al. Position statement: definition of traumatic brain injury. Arch Phys Med Rehabil. 2010;91:1637–40.PubMedCrossRef
168.
go back to reference Marion DW, Curley KC, Schwab K, Hicks RR, mTBI Diagonstics Workgroup. Proceedings of the military mTBI diagnostics workshop, St. Pete Beach, august 2010. J Neurotrauma. 2011, 28:517–26. Marion DW, Curley KC, Schwab K, Hicks RR, mTBI Diagonstics Workgroup. Proceedings of the military mTBI diagnostics workshop, St. Pete Beach, august 2010. J Neurotrauma. 2011, 28:517–26.
169.
go back to reference Teasdale G, Maas A, Lecky F, Manley G, Stocchetti N, Murray G. The Glasgow coma scale at 40 years: standing the test of time. Lancet Neurol. 2014;13:844–54.PubMedCrossRef Teasdale G, Maas A, Lecky F, Manley G, Stocchetti N, Murray G. The Glasgow coma scale at 40 years: standing the test of time. Lancet Neurol. 2014;13:844–54.PubMedCrossRef
171.
go back to reference Kulbe JR, Geddes JW. Current status of fluid biomarkers in mild traumatic brain injury. Exp Neurol. 2016;275:334–52.PubMedCrossRef Kulbe JR, Geddes JW. Current status of fluid biomarkers in mild traumatic brain injury. Exp Neurol. 2016;275:334–52.PubMedCrossRef
172.
go back to reference Townend W, Dibble C, Abid K, Vail A, Sherwood R, Lecky F. Rapid elimination of protein S-100B from serum after minor head trauma. J Neurotrauma. 2006;23:149–55.PubMedCrossRef Townend W, Dibble C, Abid K, Vail A, Sherwood R, Lecky F. Rapid elimination of protein S-100B from serum after minor head trauma. J Neurotrauma. 2006;23:149–55.PubMedCrossRef
173.
go back to reference Gatson JW, Barillas J, Hynan LS, Diaz-Arrastia R, Wolf SE, Minei JP. Detection of neurofilament-H in serum as a diagnostic tool to predict injury severity in patients who have suffered mild traumatic brain injury. J Neurosurg. 2014;121:1232–8.PubMedCrossRef Gatson JW, Barillas J, Hynan LS, Diaz-Arrastia R, Wolf SE, Minei JP. Detection of neurofilament-H in serum as a diagnostic tool to predict injury severity in patients who have suffered mild traumatic brain injury. J Neurosurg. 2014;121:1232–8.PubMedCrossRef
174.
go back to reference Papa L, Silvestri S, Brophy GM, Giordano P, Falk JL, Braga CF, et al. GFAP out-performs S100beta in detecting traumatic intracranial lesions on computed tomography in trauma patients with mild traumatic brain injury and those with extracranial lesions. J Neurotrauma. 2014;31:1815–22.PubMedPubMedCentralCrossRef Papa L, Silvestri S, Brophy GM, Giordano P, Falk JL, Braga CF, et al. GFAP out-performs S100beta in detecting traumatic intracranial lesions on computed tomography in trauma patients with mild traumatic brain injury and those with extracranial lesions. J Neurotrauma. 2014;31:1815–22.PubMedPubMedCentralCrossRef
175.
go back to reference Biberthaler P, Linsenmeier U, Pfeifer KJ, Kroetz M, Mussack T, Kanz KG, et al. Serum S-100B concentration provides additional information fot the indication of computed tomography in patients after minor head injury: a prospective multicenter study. Shock. 2006;25:446–53.PubMedCrossRef Biberthaler P, Linsenmeier U, Pfeifer KJ, Kroetz M, Mussack T, Kanz KG, et al. Serum S-100B concentration provides additional information fot the indication of computed tomography in patients after minor head injury: a prospective multicenter study. Shock. 2006;25:446–53.PubMedCrossRef
176.
go back to reference Tenovuo O, Posti J, Hossain I, Takalak R, Liedes H, Newcombe V, et al. GFAP and UCH-L1 are not specific biomarkers for mild CT-negative traumatic brain injury. Abstr. 34th Annu. Natl. Neurotrauma Symp. 2016. p. A-20. Tenovuo O, Posti J, Hossain I, Takalak R, Liedes H, Newcombe V, et al. GFAP and UCH-L1 are not specific biomarkers for mild CT-negative traumatic brain injury. Abstr. 34th Annu. Natl. Neurotrauma Symp. 2016. p. A-20.
177.
go back to reference Bazarian JJ, Zemlan FP, Mookerjee S, Stigbrand T. Serum S-100B and cleaved-tau are poor predictors of long-term outcome after mild traumatic brain injury. Brain Inj. 2006;20:759–65.PubMedCrossRef Bazarian JJ, Zemlan FP, Mookerjee S, Stigbrand T. Serum S-100B and cleaved-tau are poor predictors of long-term outcome after mild traumatic brain injury. Brain Inj. 2006;20:759–65.PubMedCrossRef
178.
go back to reference Kou Z, Gattu R, Kobeissy F, Welch RD, O’Neil BJ, Woodard JL, et al. Combining biochemical and imaging markers to improve diagnosis and characterization of mild traumatic brain injury in the acute setting: results from a pilot study. PLoS One. 2013;8:e80296.PubMedPubMedCentralCrossRef Kou Z, Gattu R, Kobeissy F, Welch RD, O’Neil BJ, Woodard JL, et al. Combining biochemical and imaging markers to improve diagnosis and characterization of mild traumatic brain injury in the acute setting: results from a pilot study. PLoS One. 2013;8:e80296.PubMedPubMedCentralCrossRef
179.
go back to reference Kavalci C, Pekdemir M, Durukan P, Ilhan N, Yildiz M, Serhatlioglu S, et al. The value of serum tau protein for the diagnosis of intracranial injury in minor head trauma. Am J Emerg Med. 2007;25:391–5.PubMedCrossRef Kavalci C, Pekdemir M, Durukan P, Ilhan N, Yildiz M, Serhatlioglu S, et al. The value of serum tau protein for the diagnosis of intracranial injury in minor head trauma. Am J Emerg Med. 2007;25:391–5.PubMedCrossRef
180.
go back to reference Luo J, Uchida K, Shi R. Accumulation of acrolein-protein adducts after traumatic spinal cord injury. Neurochem Res. 2005;30:291–5.PubMedCrossRef Luo J, Uchida K, Shi R. Accumulation of acrolein-protein adducts after traumatic spinal cord injury. Neurochem Res. 2005;30:291–5.PubMedCrossRef
181.
go back to reference Wood PL, Khan MA, Moskal JR, Todd KG, Tanay VA, Baker G. Aldehyde load in ischemia-reperfusion brain injury: neuroprotection by neutralization of reactive aldehydes with phenelzine. Brain Res. 2006;1122:184–90.PubMedCrossRef Wood PL, Khan MA, Moskal JR, Todd KG, Tanay VA, Baker G. Aldehyde load in ischemia-reperfusion brain injury: neuroprotection by neutralization of reactive aldehydes with phenelzine. Brain Res. 2006;1122:184–90.PubMedCrossRef
182.
go back to reference Putiev YP, Tashpulatov YT, Gafurov TG, Usmanov KU. Infrared study of modified cellulose. Polym Sci USSR. 1964;6:1565–70.CrossRef Putiev YP, Tashpulatov YT, Gafurov TG, Usmanov KU. Infrared study of modified cellulose. Polym Sci USSR. 1964;6:1565–70.CrossRef
183.
go back to reference Kaminskas LM, Pyke SM, Burcham PC. Strong protein adduct trapping accompanies abolition of acrolein-mediated hepatotoxicity by hydralazine in mice. J Pharmacol Exp Ther. 2004;310:1003–10.PubMedCrossRef Kaminskas LM, Pyke SM, Burcham PC. Strong protein adduct trapping accompanies abolition of acrolein-mediated hepatotoxicity by hydralazine in mice. J Pharmacol Exp Ther. 2004;310:1003–10.PubMedCrossRef
184.
go back to reference Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D. Antioxidant therapy in acute central nervous system injury: current state. Pharmacol Rev. 2002;54:271–84.PubMedCrossRef Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D. Antioxidant therapy in acute central nervous system injury: current state. Pharmacol Rev. 2002;54:271–84.PubMedCrossRef
185.
go back to reference Bains M, Hall ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta. 1822;2012:675–84. Bains M, Hall ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta. 1822;2012:675–84.
186.
go back to reference Kochanek PM, Jackson TC, Ferguson NM, Carlson SW, Simon DW, Brockman EC, et al. Emerging therapies in traumatic brain injury. Semin Neurol. 2015;35:83–100.PubMedPubMedCentralCrossRef Kochanek PM, Jackson TC, Ferguson NM, Carlson SW, Simon DW, Brockman EC, et al. Emerging therapies in traumatic brain injury. Semin Neurol. 2015;35:83–100.PubMedPubMedCentralCrossRef
187.
go back to reference Bavarsad Shahripour R, Harrigan MR, Alexandrov AV. N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities. Brain Behav. 2014;4:108–22.PubMedPubMedCentralCrossRef Bavarsad Shahripour R, Harrigan MR, Alexandrov AV. N-acetylcysteine (NAC) in neurological disorders: mechanisms of action and therapeutic opportunities. Brain Behav. 2014;4:108–22.PubMedPubMedCentralCrossRef
Metadata
Title
Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases
Authors
Marcela Cruz-Haces
Jonathan Tang
Glen Acosta
Joseph Fernandez
Riyi Shi
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2017
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-017-0088-2