Semin Neurol 2015; 35(01): 083-100
DOI: 10.1055/s-0035-1544237
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Emerging Therapies in Traumatic Brain Injury

Patrick M. Kochanek
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Travis C. Jackson
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Nikki Miller Ferguson
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Shaun W. Carlson
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
3   Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Dennis W. Simon
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Erik C. Brockman
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Jing Ji
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Hülya Bayır
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
7   Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
,
Samuel M. Poloyac
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
8   University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
,
Amy K. Wagner
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
4   Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Anthony E. Kline
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
4   Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Philip E. Empey
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
8   University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
,
Robert S.B. Clark
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
2   Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
6   Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
Edwin K. Jackson
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
5   Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
,
C. Edward Dixon
1   Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
3   Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
› Author Affiliations
Further Information

Publication History

Publication Date:
25 February 2015 (online)

Abstract

Despite decades of basic and clinical research, treatments to improve outcomes after traumatic brain injury (TBI) are limited. However, based on the recent recognition of the prevalence of mild TBI, and its potential link to neurodegenerative disease, many new and exciting secondary injury mechanisms have been identified and several new therapies are being evaluated targeting both classic and novel paradigms. This includes a robust increase in both preclinical and clinical investigations. Using a mechanism-based approach the authors define the targets and emerging therapies for TBI. They address putative new therapies for TBI across both the spectrum of injury severity and the continuum of care, from the field to rehabilitation. They discussTBI therapy using 11 categories, namely, (1) excitotoxicity and neuronal death, (2) brain edema, (3) mitochondria and oxidative stress, (4) axonal injury, (5) inflammation, (6) ischemia and cerebral blood flow dysregulation, (7) cognitive enhancement, (8) augmentation of endogenous neuroprotection, (9) cellular therapies, (10) combination therapy, and (11) TBI resuscitation. The current golden age of TBI research represents a special opportunity for the development of breakthroughs in the field.

 
  • References

  • 1 Robertson CS, Hannay HJ, Yamal JM , et al; Epo Severe TBI Trial Investigators. Effect of erythropoietin and transfusion threshold on neurological recovery after traumatic brain injury: a randomized clinical trial. JAMA 2014; 312 (1) 36-47
  • 2 Giacino JT, Whyte J, Bagiella E , et al. Placebo-controlled trial of amantadine for severe traumatic brain injury. N Engl J Med 2012; 366 (9) 819-826
  • 3 Diaz-Arrastia R, Kochanek PM, Bergold P , et al. Pharmacotherapy of traumatic brain injury: state of the science and the road forward: report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma 2014; 31 (2) 135-158
  • 4 Olney JW. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science 1969; 164 (3880) 719-721
  • 5 Ikonomidou C, Turski L. Why did NMDA receptor antagonists fail clinical trials for stroke and traumatic brain injury?. Lancet Neurol 2002; 1 (6) 383-386
  • 6 Randall RD, Thayer SA. Glutamate-induced calcium transient triggers delayed calcium overload and neurotoxicity in rat hippocampal neurons. J Neurosci 1992; 12 (5) 1882-1895
  • 7 Choi DW. Ionic dependence of glutamate neurotoxicity. J Neurosci 1987; 7 (2) 369-379
  • 8 Chamoun R, Suki D, Gopinath SP, Goodman JC, Robertson C. Role of extracellular glutamate measured by cerebral microdialysis in severe traumatic brain injury. J Neurosurg 2010; 113 (3) 564-570
  • 9 Weber JT, Rzigalinski BA, Ellis EF. Traumatic injury of cortical neurons causes changes in intracellular calcium stores and capacitative calcium influx. J Biol Chem 2001; 276 (3) 1800-1807
  • 10 Saatman KE, Bozyczko-Coyne D, Marcy V, Siman R, McIntosh TK. Prolonged calpain-mediated spectrin breakdown occurs regionally following experimental brain injury in the rat. J Neuropathol Exp Neurol 1996; 55 (7) 850-860
  • 11 Sun DA, Deshpande LS, Sombati S , et al. Traumatic brain injury causes a long-lasting calcium (Ca2+)-plateau of elevated intracellular Ca levels and altered Ca2+ homeostatic mechanisms in hippocampal neurons surviving brain injury. Eur J Neurosci 2008; 27 (7) 1659-1672
  • 12 Saatman KE, Murai H, Bartus RT , et al. Calpain inhibitor AK295 attenuates motor and cognitive deficits following experimental brain injury in the rat. Proc Natl Acad Sci U S A 1996; 93 (8) 3428-3433
  • 13 Baumgartner HK, Gerasimenko JV, Thorne C , et al. Calcium elevation in mitochondria is the main Ca2+ requirement for mitochondrial permeability transition pore (mPTP) opening. J Biol Chem 2009; 284 (31) 20796-20803
  • 14 Knoblach SM, Nikolaeva M, Huang X , et al. Multiple caspases are activated after traumatic brain injury: evidence for involvement in functional outcome. J Neurotrauma 2002; 19 (10) 1155-1170
  • 15 Conti AC, Raghupathi R, Trojanowski JQ, McIntosh TK. Experimental brain injury induces regionally distinct apoptosis during the acute and delayed post-traumatic period. J Neurosci 1998; 18 (15) 5663-5672
  • 16 Clark RS, Kochanek PM, Watkins SC , et al. Caspase-3 mediated neuronal death after traumatic brain injury in rats. J Neurochem 2000; 74 (2) 740-753
  • 17 Knoblach SM, Alroy DA, Nikolaeva M, Cernak I, Stoica BA, Faden AI. Caspase inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury. J Cereb Blood Flow Metab 2004; 24 (10) 1119-1132
  • 18 Chen W, Zhou Z, Li L , et al. Diverse sequence determinants control human and mouse receptor interacting protein 3 (RIP3) and mixed lineage kinase domain-like (MLKL) interaction in necroptotic signaling. J Biol Chem 2013; 288 (23) 16247-16261
  • 19 Kaiser WJ, Upton JW, Long AB , et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 2011; 471 (7338) 368-372
  • 20 Liu Q, Qiu J, Liang M , et al. Akt and mTOR mediate programmed necrosis in neurons. Cell Death Dis 2014; 5: e1084
  • 21 You Z, Savitz SI, Yang J , et al. Necrostatin-1 reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 2008; 28 (9) 1564-1573
  • 22 Li Y, Yang X, Ma C, Qiao J, Zhang C. Necroptosis contributes to the NMDA-induced excitotoxicity in rat's cultured cortical neurons. Neurosci Lett 2008; 447 (2-3) 120-123
  • 23 Pickford F, Masliah E, Britschgi M , et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 2008; 118 (6) 2190-2199
  • 24 Clark RS, Bayır H, Chu CT, Alber SM, Kochanek PM, Watkins SC. Autophagy is increased in mice after traumatic brain injury and is detectable in human brain after trauma and critical illness. Autophagy 2008; 4 (1) 88-90
  • 25 Luo CL, Li BX, Li QQ , et al. Autophagy is involved in traumatic brain injury-induced cell death and contributes to functional outcome deficits in mice. Neuroscience 2011; 184: 54-63
  • 26 Zhang YB, Li SX, Chen XP , et al. Autophagy is activated and might protect neurons from degeneration after traumatic brain injury. Neurosci Bull 2008; 24 (3) 143-149
  • 27 Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: effects of oligomeric and fibrillar amyloid-beta. J Neuroimmunol 2009; 210 (1-2) 3-12
  • 28 Vespa PM, Miller C, McArthur D , et al. Nonconvulsive electrographic seizures after traumatic brain injury result in a delayed, prolonged increase in intracranial pressure and metabolic crisis. Crit Care Med 2007; 35 (12) 2830-2836
  • 29 Tovar KR, Westbrook GL. The incorporation of NMDA receptors with a distinct subunit composition at nascent hippocampal synapses in vitro. J Neurosci 1999; 19 (10) 4180-4188
  • 30 Kaufman AM, Milnerwood AJ, Sepers MD , et al. Opposing roles of synaptic and extrasynaptic NMDA receptor signaling in cocultured striatal and cortical neurons. J Neurosci 2012; 32 (12) 3992-4003
  • 31 Papadia S, Soriano FX, Léveillé F , et al. Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nat Neurosci 2008; 11 (4) 476-487
  • 32 Hardingham GE, Fukunaga Y, Bading H. Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat Neurosci 2002; 5 (5) 405-414
  • 33 Karpova A, Mikhaylova M, Bera S , et al. Encoding and transducing the synaptic or extrasynaptic origin of NMDA receptor signals to the nucleus. Cell 2013; 152 (5) 1119-1133
  • 34 Bigford GE, Alonso OF, Dietrich D, Keane RW. A novel protein complex in membrane rafts linking the NR2B glutamate receptor and autophagy is disrupted following traumatic brain injury. J Neurotrauma 2009; 26 (5) 703-720
  • 35 Tu W, Xu X, Peng L , et al. DAPK1 interaction with NMDA receptor NR2B subunits mediates brain damage in stroke. Cell 2010; 140 (2) 222-234
  • 36 Ferrario CR, Ndukwe BO, Ren J, Satin LS, Goforth PB. Stretch injury selectively enhances extrasynaptic, GluN2B-containing NMDA receptor function in cortical neurons. J Neurophysiol 2013; 110 (1) 131-140
  • 37 Rao VLR, Dogan A, Todd KG, Bowen KK, Dempsey RJ. Neuroprotection by memantine, a non-competitive NMDA receptor antagonist after traumatic brain injury in rats. Brain Res 2001; 911: 96-100
  • 38 Xia P, Chen HSV, Zhang D, Lipton SA. Memantine preferentially blocks extrasynaptic over synaptic NMDA receptor currents in hippocampal autopsies. J Neurosci 2010; 30 (33) 11246-11250
  • 39 Rammes G, Danysz W, Parsons CG. Pharmacodynamics of memantine: an update. Curr Neuropharmacol 2008; 6 (1) 55-78
  • 40 Davies DJ, Crowe M, Lucas N , et al. A novel series of benzimidazole NR2B-selective NMDA receptor antagonists. Bioorg Med Chem Lett 2012; 22 (7) 2620-2623
  • 41 Niogi SN, Mukherjee P, Ghajar J , et al. Structural dissociation of attentional control and memory in adults with and without mild traumatic brain injury. Brain 2008; 131 (Pt 12) 3209-3221
  • 42 Bashir S, Vernet M, Yoo WK, Mizrahi I, Theoret H, Pascual-Leone A. Changes in cortical plasticity after mild traumatic brain injury. Restor Neurol Neurosci 2012; 30 (4) 277-282
  • 43 Schumann J, Alexandrovich GA, Biegon A, Yaka R. Inhibition of NR2B phosphorylation restores alterations in NMDA receptor expression and improves functional recovery following traumatic brain injury in mice. J Neurotrauma 2008; 25 (8) 945-957
  • 44 Kumar A, Zou L, Yuan X, Long Y, Yang K. N-methyl-D-aspartate receptors: transient loss of NR1/NR2A/NR2B subunits after traumatic brain injury in a rodent model. J Neurosci Res 2002; 67 (6) 781-786
  • 45 Szaflarski JP, Meckler JM, Szaflarski M, Shutter LA, Privitera MD, Yates SL. Levetiracetam use in critically ill patients. Neurocrit Care 2007; 7 (2) 140-147
  • 46 Lee CY, Chen CC, Liou HH. Levetiracetam inhibits glutamate transmission through presynaptic P/Q-type calcium channels on the granule cells of the dentate gyrus. Br J Pharmacol 2009; 158 (7) 1753-1762
  • 47 Wakita M, Kotani N, Kogure K, Akaike N. Inhibition of excitatory synaptic transmission in hippocampal neurons by levetiracetam involves Zn2+-dependent GABA type A receptor-mediated presynaptic modulation. J Pharmacol Exp Ther 2014; 348 (2) 246-259
  • 48 Benge JF, Phenis RA, Bernett A, Cruz-Laureano D, Kirmani BF. Neurobehavioral effects of levetiracetam in patients with traumatic brain injury. Front Neurol 2013; 4: 195
  • 49 Zou H, Brayer SW, Hurwitz M, Niyonkuru C, Fowler LE, Wagner AK. Neuroprotective, neuroplastic, and neurobehavioral effects of daily treatment with levetiracetam in experimental traumatic brain injury. Neurorehabil Neural Repair 2013; 27 (9) 878-888
  • 50 Bratton SL, Chestnut RM, Ghajar J , et al; Brain Trauma Foundation; American Association of Neurological Surgeons; Congress of Neurological Surgeons; Joint Section on Neurotrauma and Critical Care, AANS/CNS. Guidelines for the management of severe traumatic brain injury. II. Hyperosmolar therapy. J Neurotrauma 2007; 24 (Suppl. 01) S14-S20
  • 51 Kochanek PM, Carney N, Adelson PD , et al. Guidelines for the acute medical management of severe traumatic brain injury in infants, children, and adolescents – second edition. Pediatr Crit Care Med 2012; 13 (Suppl. 01) S1-82
  • 52 Gonda DD, Meltzer HS, Crawford JR , et al. Complications associated with prolonged hypertonic saline therapy in children with elevated intracranial pressure. Pediatr Crit Care Med 2013; 14 (6) 610-620
  • 53 Cooper DJ, Rosenfeld JV, Murray L , et al; DECRA Trial Investigators; Australian and New Zealand Intensive Care Society Clinical Trials Group. Decompressive craniectomy in diffuse traumatic brain injury. N Engl J Med 2011; 364 (16) 1493-1502
  • 54 Shafi S, Diaz-Arrastia R, Madden C, Gentilello L. Intracranial pressure monitoring in brain-injured patients is associated with worsening of survival. J Trauma 2008; 64 (2) 335-340
  • 55 Chesnut RM, Temkin N, Carney N , et al; Global Neurotrauma Research Group. A trial of intracranial-pressure monitoring in traumatic brain injury. N Engl J Med 2012; 367 (26) 2471-2481
  • 56 Stein DM, Hu PF, Brenner M , et al. Brief episodes of intracranial hypertension and cerebral hypoperfusion are associated with poor functional outcome after severe traumatic brain injury. J Trauma 2011; 71 (2) 364-373 , discussion 373–374
  • 57 Lafrenaye AD, McGinn MJ, Povlishock JT. Increased intracranial pressure after diffuse traumatic brain injury exacerbates neuronal somatic membrane poration but not axonal injury: evidence for primary intracranial pressure-induced neuronal perturbation. J Cereb Blood Flow Metab 2012; 32 (10) 1919-1932
  • 58 Laird MD, Shields JS, Sukumari-Ramesh S , et al. High mobility group box protein-1 promotes cerebral edema after traumatic brain injury via activation of toll-like receptor 4. Glia 2014; 62 (1) 26-38
  • 59 Badaut J, Lasbennes F, Magistretti PJ, Regli L. Aquaporins in brain: distribution, physiology, and pathophysiology. J Cereb Blood Flow Metab 2002; 22 (4) 367-378
  • 60 Bell MJ, Kochanek PM, Doughty LA , et al. Interleukin-6 and interleukin-10 in cerebrospinal fluid after severe traumatic brain injury in children. J Neurotrauma 1997; 14 (7) 451-457
  • 61 Au AK, Aneja RK, Bell MJ , et al. Cerebrospinal fluid levels of high-mobility group box 1 and cytochrome C predict outcome after pediatric traumatic brain injury. J Neurotrauma 2012; 29 (11) 2013-2021
  • 62 Okuma Y, Liu K, Wake H , et al. Glycyrrhizin inhibits traumatic brain injury by reducing HMGB1-RAGE interaction. Neuropharmacology 2014; 85: 18-26
  • 63 Simard JM, Woo SK, Schwartzbauer GT, Gerzanich V. Sulfonylurea receptor 1 in central nervous system injury: a focused review. J Cereb Blood Flow Metab 2012; 32 (9) 1699-1717
  • 64 Okuma Y, Liu K, Wake H , et al. Anti-high mobility group box-1 antibody therapy for traumatic brain injury. Ann Neurol 2012; 72 (3) 373-384
  • 65 Fukuda AM, Adami A, Pop V , et al. Posttraumatic reduction of edema with aquaporin-4 RNA interference improves acute and chronic functional recovery. J Cereb Blood Flow Metab 2013; 33 (10) 1621-1632
  • 66 Zweckberger K, Hackenberg K, Jung CS , et al. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience 2014; 272: 199-206
  • 67 Kagan VE, Wipf P, Stoyanovsky D , et al. Mitochondrial targeting of electron scavenging antioxidants: regulation of selective oxidation vs random chain reactions. Adv Drug Deliv Rev 2009; 61 (14) 1375-1385
  • 68 Tyurina YY, Poloyac SM, Tyurin VA , et al. A mitochondrial pathway for biosynthesis of lipid mediators. Nat Chem 2014; 6 (6) 542-552
  • 69 Chu CT, Ji J, Dagda RK , et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol 2013; 15 (10) 1197-1205
  • 70 Ji J, Kline AE, Amoscato A , et al. Lipidomics identifies cardiolipin oxidation as a mitochondrial target for redox therapy of brain injury. Nat Neurosci 2012; 15 (10) 1407-1413
  • 71 Okonkwo DO, Povlishock JT. An intrathecal bolus of cyclosporin A before injury preserves mitochondrial integrity and attenuates axonal disruption in traumatic brain injury. J Cereb Blood Flow Metab 1999; 19 (4) 443-451
  • 72 Xiong Y, Gu Q, Peterson PL, Muizelaar JP, Lee CP. Mitochondrial dysfunction and calcium perturbation induced by traumatic brain injury. J Neurotrauma 1997; 14 (1) 23-34
  • 73 Robertson CL. Mitochondrial dysfunction contributes to cell death following traumatic brain injury in adult and immature animals. J Bioenerg Biomembr 2004; 36 (4) 363-368
  • 74 Singh IN, Sullivan PG, Deng Y, Mbye LH, Hall ED. Time course of post-traumatic mitochondrial oxidative damage and dysfunction in a mouse model of focal traumatic brain injury: implications for neuroprotective therapy. J Cereb Blood Flow Metab 2006; 26 (11) 1407-1418
  • 75 Abdul-Muneer PM, Schuetz H, Wang F , et al. Induction of oxidative and nitrosative damage leads to cerebrovascular inflammation in an animal model of mild traumatic brain injury induced by primary blast. Free Radic Biol Med 2013; 60: 282-291
  • 76 Sies H. Oxidative stress, introductory remarks. In: Oxidative Stress. London, England: Academic Press; 1985: 1-7
  • 77 Thiels E, Urban NN, Gonzalez-Burgos GR , et al. Impairment of long-term potentiation and associative memory in mice that overexpress extracellular superoxide dismutase. J Neurosci 2000; 20 (20) 7631-7639
  • 78 Jones DP. Redefining oxidative stress. Antioxid Redox Signal 2006; 8 (9-10) 1865-1879
  • 79 Opii WO, Nukala VN, Sultana R , et al. Proteomic identification of oxidized mitochondrial proteins following experimental traumatic brain injury. J Neurotrauma 2007; 24 (5) 772-789
  • 80 Mendez DR, Cherian L, Moore N, Arora T, Liu PK, Robertson CS. Oxidative DNA lesions in a rodent model of traumatic brain injury. J Trauma 2004; 56 (6) 1235-1240
  • 81 Bayır H, Tyurin VA, Tyurina YY , et al. Selective early cardiolipin peroxidation after traumatic brain injury: an oxidative lipidomics analysis. Ann Neurol 2007; 62 (2) 154-169
  • 82 Cristofori L, Tavazzi B, Gambin R , et al. Early onset of lipid peroxidation after human traumatic brain injury: a fatal limitation for the free radical scavenger pharmacological therapy?. J Investig Med 2001; 49 (5) 450-458
  • 83 Bayır H, Kagan VE, Tyurina YY , et al. Assessment of antioxidant reserves and oxidative stress in cerebrospinal fluid after severe traumatic brain injury in infants and children. Pediatr Res 2002; 51 (5) 571-578
  • 84 Hamm RJ, Temple MD, Pike BR, Ellis EF. The effect of postinjury administration of polyethylene glycol-conjugated superoxide dismutase (pegorgotein, Dismutec) or lidocaine on behavioral function following fluid-percussion brain injury in rats. J Neurotrauma 1996; 13 (6) 325-332
  • 85 Hall ED, Andrus PK, Smith SL , et al. Pyrrolopyrimidines: novel brain-penetrating antioxidants with neuroprotective activity in brain injury and ischemia models. J Pharmacol Exp Ther 1997; 281 (2) 895-904
  • 86 Wang GH, Jiang ZL, Li YC , et al. Free-radical scavenger edaravone treatment confers neuroprotection against traumatic brain injury in rats. J Neurotrauma 2011; 28 (10) 2123-2134
  • 87 Marshall LF, Maas AI, Marshall SB , et al. A multicenter trial on the efficacy of using tirilazad mesylate in cases of head injury. J Neurosurg 1998; 89 (4) 519-525
  • 88 Muizelaar JP, Kupiec JW, Rapp LA. PEG-SOD after head injury. J Neurosurg 1995; 83 (5) 942
  • 89 Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 2014; 94 (3) 909-950
  • 90 Bayır H, Kochanek PM, Kagan VE. Oxidative stress in immature brain after traumatic brain injury. Dev Neurosci 2006; 28 (4-5) 420-431 Review
  • 91 Hall ED, Wang JA, Miller DM. Relationship of nitric oxide synthase induction to peroxynitrite-mediated oxidative damage during the first week after experimental traumatic brain injury. Exp Neurol 2012; 238 (2) 176-182
  • 92 Wang F, Franco R, Skotak M, Hu G, Chandra N. Mechanical stretch exacerbates the cell death in SH-SY5Y cells exposed to paraquat: mitochondrial dysfunction and oxidative stress. Neurotoxicology 2014; 41: 54-63
  • 93 Wu A, Ying Z, Gomez-Pinilla F. Omega-3 fatty acids supplementation restores mechanisms that maintain brain homeostasis in traumatic brain injury. J Neurotrauma 2007; 24 (10) 1587-1595
  • 94 Sullivan PG, Rabchevsky AG, Waldmeier PC, Springer JE. Mitochondrial permeability transition in CNS trauma: cause or effect of neuronal cell death?. J Neurosci Res 2005; 79 (1-2) 231-239
  • 95 Van Houten B, Woshner V, Santos JH. Role of mitochondrial DNA in toxic responses to oxidative stress. DNA Repair (Amst) 2006; 5 (2) 145-152
  • 96 Scafidi S, Racz J, Hazelton J, McKenna MC, Fiskum G. Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain. Dev Neurosci 2010; 32 (5-6) 480-487
  • 97 Scheff SW, Sullivan PG. Cyclosporin A significantly ameliorates cortical damage following experimental traumatic brain injury in rodents. J Neurotrauma 1999; 16 (9) 783-792
  • 98 Mazzeo AT, Brophy GM, Gilman CB , et al. Safety and tolerability of cyclosporin a in severe traumatic brain injury patients: results from a prospective randomized trial. J Neurotrauma 2009; 26 (12) 2195-2206
  • 99 Smith RA, Hartley RC, Murphy MP. Mitochondria-targeted small molecule therapeutics and probes. Antioxid Redox Signal 2011; 15 (12) 3021-3038
  • 100 Smith RA, Murphy MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci 2010; 1201: 96-103
  • 101 Cho J, Won K, Wu D , et al. Potent mitochondria-targeted peptides reduce myocardial infarction in rats. Coron Artery Dis 2007; 18 (3) 215-220
  • 102 Wipf P, Xiao J, Jiang J , et al. Mitochondrial targeting of selective electron scavengers: synthesis and biological analysis of hemigramicidin-TEMPO conjugates. J Am Chem Soc 2005; 127 (36) 12460-12461
  • 103 Davalos D, Grutzendler J, Yang G , et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci 2005; 8 (6) 752-758
  • 104 Shohami E, Novikov M, Bass R, Yamin A, Gallily R. Closed head injury triggers early production of TNF alpha and IL-6 by brain tissue. J Cereb Blood Flow Metab 1994; 14 (4) 615-619
  • 105 Fan L, Young PR, Barone FC, Feuerstein GZ, Smith DH, McIntosh TK. Experimental brain injury induces expression of interleukin-1 beta mRNA in the rat brain. Brain Res Mol Brain Res 1995; 30 (1) 125-130
  • 106 Aguzzi A, Barres BA, Bennett ML. Microglia: scapegoat, saboteur, or something else?. Science 2013; 339 (6116) 156-161
  • 107 Scherbel U, Raghupathi R, Nakamura M , et al. Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury. Proc Natl Acad Sci U S A 1999; 96 (15) 8721-8726
  • 108 Chio CC, Chang CH, Wang CC , et al. Etanercept attenuates traumatic brain injury in rats by reducing early microglial expression of tumor necrosis factor-α. BMC Neurosci 2013; 14: 33
  • 109 Baratz R, Tweedie D, Rubovitch V , et al. Tumor necrosis factor-α synthesis inhibitor, 3,6′-dithiothalidomide, reverses behavioral impairments induced by minimal traumatic brain injury in mice. J Neurochem 2011; 118 (6) 1032-1042
  • 110 Su X, Wang H, Zhao J, Pan H, Mao L. Beneficial effects of ethyl pyruvate through inhibiting high-mobility group box 1 expression and TLR4/NF-kenefB pathway after traumatic brain injury in the rat. Mediators Inflamm 2011; 2011: 807–142
  • 111 Zhang D, Li H, Li T , et al. TLR4 inhibitor resatorvid provides neuroprotection in experimental traumatic brain injury: implication in the treatment of human brain injury. Neurochem Int 2014; 75: 11-18
  • 112 Sanchez Mejia RO, Ona VO, Li M, Friedlander RM. Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurgery 2001; 48 (6) 1393-1399 , discussion 1399–1401
  • 113 Clausen F, Hånell A, Björk M , et al. Neutralization of interleukin-1beta modifies the inflammatory response and improves histological and cognitive outcome following traumatic brain injury in mice. Eur J Neurosci 2009; 30 (3) 385-396
  • 114 Tehranian R, Andell-Jonsson S, Beni SM , et al. Improved recovery and delayed cytokine induction after closed head injury in mice with central overexpression of the secreted isoform of the interleukin-1 receptor antagonist. J Neurotrauma 2002; 19 (8) 939-951
  • 115 Helmy A, Guilfoyle MR, Carpenter KL, Pickard JD, Menon DK, Hutchinson PJ. Recombinant human interleukin-1 receptor antagonist in severe traumatic brain injury: a phase II randomized control trial. J Cereb Blood Flow Metab 2014; 34 (5) 845-851
  • 116 Tapia-Perez J, Sanchez-Aguilar M, Torres-Corzo JG , et al. Effect of rosuvastatin on amnesia and disorientation after traumatic brain injury (NCT003229758). J Neurotrauma 2008; 25 (8) 1011-1017
  • 117 Schneider EB, Efron DT, MacKenzie EJ, Rivara FP, Nathens AB, Jurkovich GJ. Premorbid statin use is associated with improved survival and functional outcomes in older head-injured individuals. J Trauma 2011; 71 (4) 815-819
  • 118 Giunti D, Parodi B, Cordano C, Uccelli A, Kerlero de Rosbo N. Can we switch microglia's phenotype to foster neuroprotection? Focus on multiple sclerosis. Immunology 2014; 141 (3) 328-339
  • 119 Sikoglu EM, Heffernan ME, Tam K , et al. Enhancement in cognitive function recovery by granulocyte-colony stimulating factor in a rodent model of traumatic brain injury. Behav Brain Res 2014; 259: 354-356
  • 120 Acosta SA, Tajiri N, Shinozuka K , et al. Combination therapy of human umbilical cord blood cells and granulocyte colony stimulating factor reduces histopathological and motor impairments in an experimental model of chronic traumatic brain injury. PLoS ONE 2014; 9 (3) e90953
  • 121 Dela Peña I, Sanberg PR, Acosta S, Tajiri N, Lin SZ, Borlongan CV. Stem cells and G-CSF for treating neuroinflammation in traumatic brain injury: aging as a comorbidity factor. J Neurosurg Sci 2014; 58 (3) 145-149
  • 122 Povlishock JT. Pathobiology of traumatically induced axonal injury in animals and man. Ann Emerg Med 1993; 22 (6) 980-986
  • 123 Povlishock JT, Christman CW. The pathobiology of traumatically induced axonal injury in animals and humans: a review of current thoughts. J Neurotrauma 1995; 12 (4) 555-564
  • 124 Smith DH, Hicks R, Povlishock JT. Therapy development for diffuse axonal injury. J Neurotrauma 2013; 30 (5) 307-323
  • 125 Okonkwo DO, Büki A, Siman R, Povlishock JT. Cyclosporin A limits calcium-induced axonal damage following traumatic brain injury. Neuroreport 1999; 10 (2) 353-358
  • 126 Tang-Schomer MD, Patel AR, Baas PW, Smith DH. Mechanical breaking of microtubules in axons during dynamic stretch injury underlies delayed elasticity, microtubule disassembly, and axon degeneration. FASEB J 2010; 24 (5) 1401-1410
  • 127 Zhilai Z, Hui Z, Anmin J, Shaoxiong M, Bo Y, Yinhai C. A combination of taxol infusion and human umbilical cord mesenchymal stem cells transplantation for the treatment of rat spinal cord injury. Brain Res 2012; 1481: 79-89
  • 128 Mbye LH, Keles E, Tao L , et al. Kollidon VA64, a membrane-resealing agent, reduces histopathology and improves functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab 2012; 32 (3) 515-524
  • 129 Büki A, Koizumi H, Povlishock JT. Moderate posttraumatic hypothermia decreases early calpain-mediated proteolysis and concomitant cytoskeletal compromise in traumatic axonal injury. Exp Neurol 1999; 159 (1) 319-328
  • 130 Zafonte RD, Bagiella E, Ansel BM , et al. Effect of citicoline on functional and cognitive status among patients with traumatic brain injury: Citicoline Brain Injury Treatment Trial (COBRIT). JAMA 2012; 308 (19) 1993-2000
  • 131 Patel SP, Sullivan PG, Pandya JD , et al. N-acetylcysteine amide preserves mitochondrial bioenergetics and improves functional recovery following spinal trauma. Exp Neurol 2014; 257: 95-105
  • 132 Clifton GL, Valadka A, Zygun D , et al. Very early hypothermia induction in patients with severe brain injury (the National Acute Brain Injury Study: Hypothermia II): a randomised trial. Lancet Neurol 2011; 10 (2) 131-139
  • 133 Su E, Bell MJ, Kochanek PM , et al. Increased CSF concentrations of myelin basic protein after TBI in infants and children: absence of significant effect of therapeutic hypothermia. Neurocrit Care 2012; 17 (3) 401-407
  • 134 Miyauchi T, Wei EP, Povlishock JT. Evidence for the therapeutic efficacy of either mild hypothermia or oxygen radical scavengers after repetitive mild traumatic brain injury. J Neurotrauma 2014; 31 (8) 773-781
  • 135 Reeves TM, Phillips LL, Lee NN, Povlishock JT. Preferential neuroprotective effect of tacrolimus (FK506) on unmyelinated axons following traumatic brain injury. Brain Res 2007; 1154: 225-236
  • 136 Cantu RC, Gean AD. Second-impact syndrome and a small subdural hematoma: an uncommon catastrophic result of repetitive head injury with a characteristic imaging appearance. J Neurotrauma 2010; 27 (9) 1557-1564
  • 137 Bouma GJ, Muizelaar JP, Stringer WA, Choi SC, Fatouros P, Young HF. Ultra-early evaluation of regional cerebral blood flow in severely head-injured patients using xenon-enhanced computerized tomography. J Neurosurg 1992; 77 (3) 360-368
  • 138 Hall CN, Reynell C, Gesslein B , et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 2014; 508 (7494) 55-60
  • 139 Terpolilli NA, Kim SW, Thal SC, Kuebler WM, Plesnila N. Inhaled nitric oxide reduces secondary brain damage after traumatic brain injury in mice. J Cereb Blood Flow Metab 2013; 33 (2) 311-318
  • 140 Armstead WM, Kiessling JW, Kofke WA, Vavilala MS. SNP improves cerebral hemodynamics during normotension but fails to prevent sex dependent impaired cerebral autoregulation during hypotension after brain injury. Brain Res 2010; 1330: 142-150
  • 141 Sinz EH, Kochanek PM, Dixon CE , et al. Inducible nitric oxide synthase is an endogenous neuroprotectant after traumatic brain injury in rats and mice. J Clin Invest 1999; 104 (5) 647-656
  • 142 Salonia R, Empey PE, Poloyac SM , et al. Endothelin-1 is increased in cerebrospinal fluid and associated with unfavorable outcomes in children after severe traumatic brain injury. J Neurotrauma 2010; 27 (10) 1819-1825
  • 143 Macdonald RL, Higashida RT, Keller E , et al. Clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid haemorrhage undergoing surgical clipping: a randomised, double-blind, placebo-controlled phase 3 trial (CONSCIOUS-2). Lancet Neurol 2011; 10 (7) 618-625
  • 144 Giannopoulos S, Katsanos AH, Tsivgoulis G, Marshall RS. Statins and cerebral hemodynamics. J Cereb Blood Flow Metab 2012; 32 (11) 1973-1976
  • 145 Jansen JO, Lord JM, Thickett DR, Midwinter MJ, McAuley DF, Gao F. Clinical review: statins and trauma—a systematic review. Crit Care 2013; 17 (3) 227
  • 146 Elkind MS, Sacco RL, MacArthur RB , et al. The Neuroprotection with Statin Therapy for Acute Recovery Trial (NeuSTART): an adaptive design phase I dose-escalation study of high-dose lovastatin in acute ischemic stroke. Int J Stroke 2008; 3 (3) 210-218
  • 147 Shaik JS, Ahmad M, Li W , et al. Soluble epoxide hydrolase inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid is neuroprotective in rat model of ischemic stroke. Am J Physiol Heart Circ Physiol 2013; 305 (11) H1605-H1613
  • 148 Poloyac SM, Zhang Y, Bies RR, Kochanek PM, Graham SH. Protective effect of the 20-HETE inhibitor HET0016 on brain damage after temporary focal ischemia. J Cereb Blood Flow Metab 2006; 26 (12) 1551-1561
  • 149 Fordsmann JC, Ko RW, Choi HB , et al. Increased 20-HETE synthesis explains reduced cerebral blood flow but not impaired neurovascular coupling after cortical spreading depression in rat cerebral cortex. J Neurosci 2013; 33 (6) 2562-2570
  • 150 Hartings JA, Bullock MR, Okonkwo DO , et al; Co-Operative Study on Brain Injury Depolarisations. Spreading depolarisations and outcome after traumatic brain injury: a prospective observational study. Lancet Neurol 2011; 10 (12) 1058-1064
  • 151 Feeney DM, Gonzalez A, Law WA. Amphetamine, haloperidol, and experience interact to affect rate of recovery after motor cortex injury. Science 1982; 217 (4562) 855-857
  • 152 Hovda DA, Sutton RL, Feeney DM. Amphetamine-induced recovery of visual cliff performance after bilateral visual cortex ablation in cats: measurements of depth perception thresholds. Behav Neurosci 1989; 103 (3) 574-584
  • 153 Feeney DM. Pharmacologic modulation of recovery after brain injury: a reconsideration of diaschisis. J Neurol Rehabil 1991; 5: 113-128
  • 154 Kline AE, Chen MJ, Tso-Olivas DY, Feeney DM. Methylphenidate treatment following ablation-induced hemiplegia in rat: experience during drug action alters effects on recovery of function. Pharmacol Biochem Behav 1994; 48 (3) 773-779
  • 155 Wagner AK, Ren D, Conley YP , et al. Sex and genetic associations with cerebrospinal fluid dopamine and metabolite production after severe traumatic brain injury. J Neurosurg 2007; 106 (4) 538-547
  • 156 Wagner AK, Scanlon JM, Becker CR , et al. The influence of genetic variations on striatal dopamine transporter and D2 receptor binding after TBI. J Cereb Blood Flow Metab 2014; 34 (8) 1328-1339
  • 157 Bales JW, Kline AE, Wagner AK, Dixon CE. Targeting dopamine acutely in traumatic brain injury. Open Drug Discov J 2010; 2: 119-128
  • 158 Dixon CE, Bales J, Kline AE, Wagner AK. Dopamine mechanisms of injury and recovery after TBI. In: Kobaissy FH, , ed. Brain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects in Brain Injury Models. Boca Raton, FL: CRC Press; 2015. ; In press
  • 159 Kline AE, Yan HQ, Bao J, Marion DW, Dixon CE. Chronic methylphenidate treatment enhances water maze performance following traumatic brain injury in rats. Neurosci Lett 2000; 280 (3) 163-166
  • 160 Wagner AK, Drewencki LL, Chen X , et al. Chronic methylphenidate treatment enhances striatal dopamine neurotransmission after experimental traumatic brain injury. J Neurochem 2009; b 108 (4) 986-997
  • 161 Wagner AK, Kline AE, Ren D , et al. Gender associations with chronic methylphenidate treatment and behavioral performance following experimental traumatic brain injury. Behav Brain Res 2007; 181 (2) 200-209
  • 162 Meythaler JM, Brunner RC, Johnson A, Novack TA. Amantadine to improve neurorecovery in traumatic brain injury-associated diffuse axonal injury: a pilot double-blind randomized trial. J Head Trauma Rehabil 2002; 17 (4) 300-313
  • 163 Dixon CE, Kraus MF, Kline AE , et al. Amantadine improves water maze performance without affecting motor behavior following traumatic brain injury in rats. Restor Neurol Neurosci 1999; 14 (4) 285-294
  • 164 Wang T, Huang XJ, Van KC, Went GT, Nguyen JT, Lyeth BG. Amantadine improves cognitive outcome and increases neuronal survival after fluid percussion traumatic brain injury in rats. J Neurotrauma 2014; 31 (4) 370-377
  • 165 Grelak RP, Clark R, Stump JM, Vernier VG. Amantadine-dopamine interaction: possible mode of action in Parkinsonism. Science 1970; 169 (3941) 203-204
  • 166 Gianutsos G, Chute S, Dunn JP. Pharmacological changes in dopaminergic systems induced by long-term administration of amantadine. Eur J Pharmacol 1985; 110 (3) 357-361
  • 167 Allen RM. Role of amantadine in the management of neuroleptic-induced extrapyramidal syndromes: overview and pharmacology. Clin Neuropharmacol 1983; 6 (Suppl. 01) S64-S73
  • 168 Harun R, Wagner AK. The neurobiological basis of pharmacological approaches for patients with traumatic brain injury. In: Levin HS, Shum DHK, Chan RCK. Understanding Traumatic Brain Injury: Current Research and Future Directions. New York, NY: Oxford University Press; 2013: 255-300
  • 169 Kline AE, Massucci JL, Ma X, Zafonte RD, Dixon CE. Bromocriptine reduces lipid peroxidation and enhances spatial learning and hippocampal neuron survival in a rodent model of focal brain trauma. J Neurotrauma 2004; 21 (12) 1712-1722
  • 170 Brannan T, Martínez-Tica J, Di Rocco A, Yahr MD. Low and high dose bromocriptine have different effects on striatal dopamine release: an in vivo study. J Neural Transm Park Dis Dement Sect 1993; 6 (2) 81-87
  • 171 Zhu J, Hamm RJ, Reeves TM, Povlishock JT, Phillips LL. Postinjury administration of L-deprenyl improves cognitive function and enhances neuroplasticity after traumatic brain injury. Exp Neurol 2000; 166 (1) 136-152
  • 172 Kraus MF, Maki PM. Effect of amantadine hydrochloride on symptoms of frontal lobe dysfunction in brain injury: case studies and review. J Neuropsychiatry Clin Neurosci 1997; 9 (2) 222-230
  • 173 McDowell S, Whyte J, D'Esposito M. Differential effect of a dopaminergic agonist on prefrontal function in traumatic brain injury patients. Brain 1998; 121 (Pt 6) 1155-1164
  • 174 Verrier JD, Jackson TC, Bansal R , et al. The brain in vivo expresses the 2′,3′-cAMP-adenosine pathway. J Neurochem 2012; 122 (1) 115-125
  • 175 Kochanek PM, Verrier JD, Wagner AK, Jackson EK. The many roles of adenosine in traumatic brain injury. In: Boison D, Masino S, , eds. Adenosine: A Key Link Between Metabolism and Central Nervous System Activity. New York, NY: Springer; 2012: 307-322
  • 176 Kochanek PM, Vagni VA, Janesko KL , et al. Adenosine A1 receptor knockout mice develop lethal status epilepticus after experimental traumatic brain injury. J Cereb Blood Flow Metab 2006; 26 (4) 565-575
  • 177 Wagner AK, Miller MA, Scanlon J, Ren D, Kochanek PM, Conley YP. Adenosine A1 receptor gene variants associated with post-traumatic seizures after severe TBI. Epilepsy Res 2010; 90 (3) 259-272
  • 178 Haselkorn ML, Shellington DK, Jackson EK , et al. Adenosine A1 receptor activation as a brake on the microglial response after experimental traumatic brain injury in mice. J Neurotrauma 2010; 27 (5) 901-910
  • 179 Kim M, Ham A, Kim KY-M, Brown KM, Lee HT. The volatile anesthetic isoflurane increases endothelial adenosine generation via microparticle ecto-5′-nucleotidase (CD73) release. PLoS ONE 2014; 9 (6) e99950
  • 180 Tas PWL, Eisemann C, Roewer N. The volatile anesthetic isoflurane suppresses spontaneous calcium oscillations in vitro in rat hippocampal neurons by activation of adenosine A1 receptors. Neurosci Lett 2003; 338 (3) 229-232
  • 181 Sachse KT, Jackson EK, Wisniewski SR , et al. Increases in cerebrospinal fluid caffeine concentration are associated with favorable outcome after severe traumatic brain injury in humans. J Cereb Blood Flow Metab 2008; 28 (2) 395-401
  • 182 Boison D. The adenosine kinase hypothesis of epileptogenesis. Prog Neurobiol 2008; 84 (3) 249-262
  • 183 Huber A, Padrun V, Déglon N, Aebischer P, Möhler H, Boison D. Grafts of adenosine-releasing cells suppress seizures in kindling epilepsy. Proc Natl Acad Sci U S A 2001; 98 (13) 7611-7616
  • 184 Baroncelli L, Braschi C, Spolidoro M, Begenisic T, Sale A, Maffei L. Nurturing brain plasticity: impact of environmental enrichment. Cell Death Differ 2010; 17 (7) 1092-1103
  • 185 Rassaf T, Totzeck M, Hendgen-Cotta UB, Shiva S, Heusch G, Kelm M. Circulating nitrite contributes to cardioprotection by remote ischemic preconditioning. Circ Res 2014; 114 (10) 1601-1610
  • 186 Dezfulian C, Shiva S, Alekseyenko A , et al. Nitrite therapy after cardiac arrest reduces reactive oxygen species generation, improves cardiac and neurological function, and enhances survival via reversible inhibition of mitochondrial complex I. Circulation 2009; 120 (10) 897-905
  • 187 Schwarzschild MA, Ascherio A, Beal MF , et al; Parkinson Study Group SURE-PD Investigators. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol 2014; 71 (2) 141-150
  • 188 Chip S, Zelmer A, Ogunshola OO , et al. The RNA-binding protein RBM3 is involved in hypothermia induced neuroprotection. Neurobiol Dis 2011; 43 (2) 388-396
  • 189 Jackson TC, Verrier JD, Kochanek PM. Anthraquinone-2-sulfonic acid (AQ2S) is a novel neurotherapeutic agent. Cell Death Dis 2013; 4: e451
  • 190 Mahmood A, Lu D, Yi L, Chen JL, Chopp M. Intracranial bone marrow transplantation after traumatic brain injury improving functional outcome in adult rats. J Neurosurg 2001; 94 (4) 589-595
  • 191 Lu D, Li Y, Wang L, Chen J, Mahmood A, Chopp M. Intraarterial administration of marrow stromal cells in a rat model of traumatic brain injury. J Neurotrauma 2001; 18 (8) 813-819
  • 192 Mahmood A, Lu D, Qu C, Goussev A, Chopp M. Long-term recovery after bone marrow stromal cell treatment of traumatic brain injury in rats. J Neurosurg 2006; 104 (2) 272-277
  • 193 Lu D, Mahmood A, Wang L, Li Y, Lu M, Chopp M. Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome. NeuroReport 2001; 12 (3) 559-563
  • 194 Riess P, Zhang C, Saatman KE , et al. Transplanted neural stem cells survive, differentiate, and improve neurological motor function after experimental traumatic brain injury. Neurosurgery 2002; 51 (4) 1043-1052 , discussion 1052–1054
  • 195 Hoane MR, Becerra GD, Shank JE , et al. Transplantation of neuronal and glial precursors dramatically improves sensorimotor function but not cognitive function in the traumatically injured brain. J Neurotrauma 2004; 21 (2) 163-174
  • 196 Liu SJ, Zou Y, Belegu V , et al. Co-grafting of neural stem cells with olfactory en sheathing cells promotes neuronal restoration in traumatic brain injury with an anti-inflammatory mechanism. J Neuroinflammation 2014; 11: 66
  • 197 Mahmood A, Wu H, Qu C, Xiong Y, Chopp M. Effects of treating traumatic brain injury with collagen scaffolds and human bone marrow stromal cells on sprouting of corticospinal tract axons into the denervated side of the spinal cord. J Neurosurg 2013; 118 (2) 381-389
  • 198 Tajiri N, Acosta SA, Shahaduzzaman M , et al. Intravenous transplants of human adipose-derived stem cell protect the brain from traumatic brain injury-induced neurodegeneration and motor and cognitive impairments: cell graft biodistribution and soluble factors in young and aged rats. J Neurosci 2014; 34 (1) 313-326
  • 199 Sun D, Bullock MR, McGinn MJ , et al. Basic fibroblast growth factor-enhanced neurogenesis contributes to cognitive recovery in rats following traumatic brain injury. Exp Neurol 2009; 216 (1) 56-65
  • 200 Kleindienst A, McGinn MJ, Harvey HB, Colello RJ, Hamm RJ, Bullock MR. Enhanced hippocampal neurogenesis by intraventricular S100B infusion is associated with improved cognitive recovery after traumatic brain injury. J Neurotrauma 2005; 22 (6) 645-655
  • 201 Bregy A, Nixon R, Lotocki G , et al. Posttraumatic hypothermia increases doublecortin expressing neurons in the dentate gyrus after traumatic brain injury in the rat. Exp Neurol 2012; 233 (2) 821-828
  • 202 Carlson SW, Madathil SK, Sama DM, Gao X, Chen J, Saatman KE. Conditional overexpression of insulin-like growth factor-1 enhances hippocampal neurogenesis and restores immature neuron dendritic processes after traumatic brain injury. J Neuropathol Exp Neurol 2014; 73 (8) 734-746
  • 203 Deng W, Aimone JB, Gage FH. New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory?. Nat Rev Neurosci 2010; 11 (5) 339-350
  • 204 Blaya MO, Bramlett HM, Naidoo J, Pieper AA, Dietrich WD. Neuroprotective efficacy of a proneurogenic compound after traumatic brain injury. J Neurotrauma 2014; 31 (5) 476-486
  • 205 Loane DJ, Faden AI. Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies. Trends Pharmacol Sci 2010; 31 (12) 596-604
  • 206 Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here?. Br J Pharmacol 2011; 164 (4) 1207-1229
  • 207 Margulies S, Hicks R ; Combination Therapies for Traumatic Brain Injury Workshop Leaders. Combination therapies for traumatic brain injury: prospective considerations. J Neurotrauma 2009; 26 (6) 925-939
  • 208 Eakin K, Baratz-Goldstein R, Pick CG , et al. Efficacy of N-acetyl cysteine in traumatic brain injury. PLoS ONE 2014; 9 (4) e90617
  • 209 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 (1) e54163
  • 210 Samuni Y, Goldstein S, Dean OM, Berk M. The chemistry and biological activities of N-acetylcysteine. Biochim Biophys Acta 2013; 1830 (8) 4117-4129
  • 211 Abdel Baki SG, Schwab B, Haber M, Fenton AA, Bergold PJ. Minocycline synergizes with N-acetylcysteine and improves cognition and memory following traumatic brain injury in rats. PLoS ONE 2010; 5 (8) e12490
  • 212 Haber M, Abdel Baki SG, Grin'kina NM , et al. Minocycline plus N-acetylcysteine synergize to modulate inflammation and prevent cognitive and memory deficits in a rat model of mild traumatic brain injury. Exp Neurol 2013; 249: 169-177
  • 213 Empey PE, Alexander HL, Ocque AJ , et al. Probenecid increases n-acetylcysteine brain penetration following experimental pediatric traumatic brain injury. J Neurotrauma 2012; 29: 229-230
  • 214 Tang H, Hua F, Wang J , et al. Progesterone and vitamin D: improvement after traumatic brain injury in middle-aged rats. Horm Behav 2013; 64 (3) 527-538
  • 215 Shear DA, Tortella FC. A military-centered approach to neuroprotection for traumatic brain injury. Front Neurol 2013; 4: 73
  • 216 Chesnut RM. Secondary brain insults after head injury: clinical perspectives. New Horiz 1995; 3 (3) 366-375
  • 217 Jungner M, Grände PO, Mattiasson G, Bentzer P. Effects on brain edema of crystalloid and albumin fluid resuscitation after brain trauma and hemorrhage in the rat. Anesthesiology 2010; 112 (5) 1194-1203
  • 218 Myburgh J, Cooper DJ, Finfer S , et al; SAFE Study Investigators; Australian and New Zealand Intensive Care Society Clinical Trials Group; Australian Red Cross Blood Service; George Institute for International Health. Saline or albumin for fluid resuscitation in patients with traumatic brain injury. N Engl J Med 2007; 357 (9) 874-884
  • 219 Chodobski A, Zink BJ, Szmydynger-Chodobska J. Blood–brain barrier pathophysiology in traumatic brain injury. Transl Stroke Res 2011; 2 (4) 492-516
  • 220 Brockman EC, Bayır H, Blasiole B , et al. Polynitroxylated-pegylated hemoglobin attenuates fluid requirements and brain edema in combined traumatic brain injury plus hemorrhagic shock in mice. J Cereb Blood Flow Metab 2013; 33 (9) 1457-1464
  • 221 Shellington DK, Du L, Wu X , et al. Polynitroxylated pegylated hemoglobin: a novel neuroprotective hemoglobin for acute volume-limited fluid resuscitation after combined traumatic brain injury and hemorrhagic hypotension in mice. Crit Care Med 2011; 39 (3) 494-505
  • 222 Brands J, Kliner D, Lipowsky HH, Kameneva MV, Villanueva FS, Pacella JJ. New insights into the microvascular mechanisms of drag reducing polymers: effect on the cell-free layer. PLoS ONE 2013; 8 (10) e77252
  • 223 Pacella JJ, Kameneva MV, Brands J , et al. Modulation of pre-capillary arteriolar pressure with drag-reducing polymers: a novel method for enhancing microvascular perfusion. Microcirculation 2012; 19 (7) 580-585
  • 224 Macias CA, Kameneva MV, Tenhunen JJ, Puyana JC, Fink MP. Survival in a rat model of lethal hemorrhagic shock is prolonged following resuscitation with a small volume of a solution containing a drag-reducing polymer derived from aloe vera. Shock 2004; 22 (2) 151-156
  • 225 Khuman J, Zhang J, Park J, Carroll JD, Donahue C, Whalen MJ. Low-level laser light therapy improves cognitive deficits and inhibits microglial activation after controlled cortical impact in mice. J Neurotrauma 2012; 29 (2) 408-417
  • 226 Singleton RH, Yan HQ, Fellows-Mayle W, Dixon CE. Resveratrol attenuates behavioral impairments and reduces cortical and hippocampal loss in a rat controlled cortical impact model of traumatic brain injury. J Neurotrauma 2010; 27 (6) 1091-1099
  • 227 Wu A, Ying Z, Gomez-Pinilla F. The salutary effects of DHA dietary supplementation on cognition, neuroplasticity, and membrane homeostasis after brain trauma. J Neurotrauma 2011; 28 (10) 2113-2122
  • 228 Dash PK, Johnson D, Clark J , et al. Involvement of the glycogen synthase kinase-3 signaling pathway in TBI pathology and neurocognitive outcome. PLoS ONE 2011; 6 (9) e24648
  • 229 Valle EJ, Allen CJ, Van Haren RM , et al. Do all trauma patients benefit from tranexamic acid?. J Trauma Acute Care Surg 2014; 76 (6) 1373-1378
  • 230 Kabadi SV, Stoica BA, Loane DJ, Luo T, Faden AI. CR8, a novel inhibitor of CDK, limits microglial activation, astrocytosis, neuronal loss, and neurologic dysfunction after experimental traumatic brain injury. J Cereb Blood Flow Metab 2014; 34 (3) 502-513
  • 231 Maruta J, Heaton KJ, Kryskow EM, Maule AL, Ghajar J. Dynamic visuomotor synchronization: quantification of predictive timing. Behav Res Methods 2013; 45 (1) 289-300