Skip to main content

Advertisement

Log in

Interactions of Oxidative Stress and Neurovascular Inflammation in the Pathogenesis of Traumatic Brain Injury

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Traumatic brain injury (TBI) is a major cause of death in the young age group and leads to persisting neurological impairment in many of its victims. It may result in permanent functional deficits because of both primary and secondary damages. This review addresses the role of oxidative stress in TBI-mediated secondary damages by affecting the function of the vascular unit, changes in blood-brain barrier (BBB) permeability, posttraumatic edema formation, and modulation of various pathophysiological factors such as inflammatory factors and enzymes associated with trauma. Oxidative stress plays a major role in many pathophysiologic changes that occur after TBI. In fact, oxidative stress occurs when there is an impairment or inability to balance antioxidant production with reactive oxygen species (ROS) and reactive nitrogen species (RNS) levels. ROS directly downregulate proteins of tight junctions and indirectly activate matrix metalloproteinases (MMPs) that contribute to open the BBB. Loosening of the vasculature and perivascular unit by oxidative stress-induced activation of MMPs and fluid channel aquaporins promotes vascular or cellular fluid edema, enhances leakiness of the BBB, and leads to progression of neuroinflammation. Likewise, oxidative stress activates directly the inflammatory cytokines and growth factors such as IL-1β, tumor necrosis factor-α (TNF-α), and transforming growth factor-beta (TGF-β) or indirectly by activating MMPs. In another pathway, oxidative stress-induced degradation of endothelial vascular endothelial growth factor receptor-2 (VEGFR-2) by MMPs leads to a subsequent elevation of cellular/serum VEGF level. The decrease in VEGFR-2 with a subsequent increase in VEGF-A level leads to apoptosis and neuroinflammation via the activation of caspase-1/3 and IL-1β release.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Lee M, Longoria R, Wilson D (1997) Ballistic waves in high-speed water entry. Fluid Struct 11(7):819–844

    Google Scholar 

  2. Teasdale G, Jennett B (1974) Assessment of coma and impaired consciousness. A practical scale. Lancet 2(7872):81–84

    CAS  PubMed  Google Scholar 

  3. Gaetz M (2004) The neurophysiology of brain injury. Clin Neurophysiol 115(1):4–18

    CAS  PubMed  Google Scholar 

  4. Maas AI, Stocchetti N, Bullock R (2008) Moderate and severe traumatic brain injury in adults. Lancet Neurol 7(8):728–741

    PubMed  Google Scholar 

  5. Beauchamp K, Mutlak H, Smith WR, Shohami E, Stahel PF (2008) Pharmacology of traumatic brain injury: where is the “golden bullet”? Mol Med 14(11–12):731–740

    PubMed Central  CAS  PubMed  Google Scholar 

  6. Lotocki G, de Rivero Vaccari JP, Perez ER, Sanchez-Molano J, Furones-Alonso O, Bramlett HM, Dietrich WD (2009) Alterations in blood-brain barrier permeability to large and small molecules and leukocyte accumulation after traumatic brain injury: effects of post-traumatic hypothermia. J Neurotrauma 26(7):1123–1134

    PubMed Central  PubMed  Google Scholar 

  7. Pun PB, Lu J, Moochhala S (2009) Involvement of ROS in BBB dysfunction. Free Radic Res 43(4):348–364

    CAS  PubMed  Google Scholar 

  8. Toklu HZ, Hakan T, Biber N, Solakoğlu S, Oğünç AV, Sener G (2009) The protective effect of alpha lipoic acid against traumatic brain injury in rats. Free Radic Res 43(7):658–667

    CAS  PubMed  Google Scholar 

  9. Abdul-Muneer PM, Schuetz H, Wang F, Skotak M, Jones J, Gorantla S, Zimmerman MC, Chandra N, Haorah J (2013) 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 60:282–291

    PubMed Central  CAS  PubMed  Google Scholar 

  10. Readnower RD, Chavko M, Adeeb S, Conroy MD, Pauly JR, McCarron RM, Sullivan PG (2010) Increase in blood-brain barrier permeability, oxidative stress, and activated microglia in a rat model of blast-induced traumatic brain injury. J Neurosci Res 88(16):3530–3539

    PubMed Central  CAS  PubMed  Google Scholar 

  11. Zhang QG, Laird MD, Han D, Nguyen K, Scott E, Dong Y, Dhandapani KM, Brann DW (2012) Critical role of NADPH oxidase in neuronal oxidative damage and microglia activation following traumatic brain injury. PLoS One 7(4):e34504

    PubMed Central  CAS  PubMed  Google Scholar 

  12. Poon HF, Calabrese V, Scapagnini G, Butterfield DA (2004) Free radicals and brain aging. Clin Geriatr Med 20(2):329–359

    PubMed  Google Scholar 

  13. Cornelius C, Crupi R, Calabrese V, Graziano A, Milone P, Pennisi G, Radak Z, Calabrese EJ, Cuzzocrea S (2013) Traumatic brain injury: oxidative stress and neuroprotection. Antioxid Redox Signal 19(8):836–853

    CAS  PubMed  Google Scholar 

  14. Herrmann M, Curio N, Jost S, Wunderlich MT, Synowitz H, Wallesch CW (1999) Protein S-100B and neuron specific enolase as early neurobiochemical markers of the severity of traumatic brain injury. Restor Neurol Neurosci 14(2–3):109–114

    CAS  Google Scholar 

  15. Berger RP, Adelson PD, Pierce MC, Dulani T, Cassidy LD, Kochanek PM (2005) Serum neuron-specific enolase, S100B, and myelin basic protein concentrations after inflicted and noninflicted traumatic brain injury in children. J Neurosurg 103(1 Suppl):61–68

    PubMed  Google Scholar 

  16. Geyer C, Ulrich A, Gräfe G, Stach B, Till H (2009) Diagnostic value of S100B and neuron-specific enolase in mild pediatric traumatic brain injury. J Neurosurg Pediatr 4(4):339–344

    PubMed  Google Scholar 

  17. de Kruijk JR, Leffers P, Menheere PP, Meerhoff S, Twijnstra A (2001) S-100B and neuron-specific enolase in serum of mild traumatic brain injury patients. A comparison with health controls. Acta Neurol Scand 103(3):175–179

    PubMed  Google Scholar 

  18. Büttner T, Weyers S, Postert T, Sprengelmeyer R, Kuhn W (1997) S-100 protein: serum marker of focal brain damage after ischemic territorial MCA infarction. Stroke 28(10):1961–1965

    PubMed  Google Scholar 

  19. Gabbita SP, Scheff SW, Menard RM, Roberts K, Fugaccia I, Zemlan FP (2005) Cleaved-tau: a biomarker of neuronal damage after traumatic brain injury. J Neurotrauma 22:83–94

    PubMed  Google Scholar 

  20. Marklund N, Blennow K, Zetterberg H, Ronne-Engström E, Enblad P, Hillered L (2009) Monitoring of brain interstitial total tau and beta amyloid proteins by microdialysis in patients with traumatic brain injury. J Neurosurg 110(6):1227–1237

    CAS  PubMed  Google Scholar 

  21. Bulut M, Koksal O, Dogan S, Bolca N, Ozguc H, Korfali E, Ilcol YO, Parklak M (2006) Tau protein as a serum marker of brain damage in mild traumatic brain injury: preliminary results. Adv Ther 23(1):12–22

    CAS  PubMed  Google Scholar 

  22. Zemlan FP, Rosenberg WS, Luebbe PA, Campbell TA, Dean GE, Weiner NE, Cohen JA, Rudick RA, Woo D (1999) Quantification of axonal damage in traumatic brain injury: affinity purification and characterization of cerebrospinal fluid tau proteins. J Neurochem 72(2):741–750

    CAS  PubMed  Google Scholar 

  23. Bazarian JJ, Zemlan FP, Mookerjee S, Stigbrand T (2006) Serum S-100B and cleaved-tau are poor predictors of long-term outcome after mild traumatic brain injury. Brain Inj 20(7):759–765

    PubMed  Google Scholar 

  24. Goldstein LE, Fisher AM, Tagge CA, Zhang XL, Velisek L, Sullivan JA, Upreti C, Kracht JM, Ericsson M, Wojnarowicz MW et al (2012) Chronic traumatic encephalopathy in blast-exposed military veterans and a blast neurotrauma mouse model. Sci Transl Med 4(134):134–160

    Google Scholar 

  25. Goedert M (2001) The significance of tau and alpha-synuclein inclusions in neurodegenerative diseases. Curr Opin Genet Dev 11(3):343–351

    CAS  PubMed  Google Scholar 

  26. Hergenroeder GW, Redell JB, Moore AN, Dash PK (2008) Biomarkers in the clinical diagnosis and management of traumatic brain injury. Mol Diagn Ther 12(6):345–358

    CAS  PubMed  Google Scholar 

  27. Balazy M, Nigam S (2003) Aging, lipid modifications and phospholipases—new concepts. Ageing Res Rev 2(2):191–209

    CAS  PubMed  Google Scholar 

  28. Brandes RP, Weissmann N, Schröder K (2010) NADPH oxidases in cardiovascular disease. Free Radic Biol Med 49(5):687–706

    CAS  PubMed  Google Scholar 

  29. Uttara B, Singh AV, Zamboni P, Mahajan RT (2009) Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol 7(1):65–74

    PubMed Central  CAS  PubMed  Google Scholar 

  30. Bedard K, Krause KH (2007) The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87(1):245–313

    CAS  PubMed  Google Scholar 

  31. Andrew PJ, Mayer B (1999) Enzymatic function of nitric oxide synthases. Cardiovasc Res 43(3):521–531

    CAS  PubMed  Google Scholar 

  32. Sastre J, Pallardó FV, Viña J (2003) The role of mitochondrial oxidative stress in aging. Free Radic Biol Med 35(1):1–8

    CAS  PubMed  Google Scholar 

  33. Choi BY, Jang BG, Kim JH, Lee BE, Sohn M, Song HK, Suh SW (2012) Prevention of traumatic brain injury-induced neuronal death by inhibition of NADPH oxidase activation. Brain Res 1481:49–58

    CAS  PubMed  Google Scholar 

  34. Shi Q, Gibson GE (2007) Oxidative stress and transcriptional regulation in Alzheimer disease. Alzheimer Dis Assoc Disord 21(4):276–291

    PubMed Central  CAS  PubMed  Google Scholar 

  35. DeWitt DS, Prough DS (2009) Blast-induced brain injury and posttraumatic hypotension and hypoxemia. J Neurotrauma 26(6):877–887

    PubMed  Google Scholar 

  36. Gahm C, Holmin S, Wiklund PN, Brundin L, Mathiesen T (2006) Neuroprotection by selective inhibition of inducible nitric oxide synthase after experimental brain contusion. J Neurotrauma 23(9):1343–1354

    PubMed  Google Scholar 

  37. Hortobágyi T, Görlach C, Benyó Z, Lacza Z, Hortobágyi S, Wahl M, Harkany T (2003) Inhibition of neuronal nitric oxide synthase-mediated activation of poly(ADP-ribose) polymerase in traumatic brain injury: neuroprotection by 3-aminobenzamide. Neuroscience 121(4):983–990

    PubMed  Google Scholar 

  38. Sinz EH, Kochanek PM, Dixon CE, Clark RS, Carcillo JA, Schiding JK, Chen M, Wisniewski SR, Carlos TM, Williams D et al (1999) Inducible nitric oxide synthase is an endogenous neuroprotectant after traumatic brain injury in rats and mice. J Clin Invest 104(5):647–656

    PubMed Central  CAS  PubMed  Google Scholar 

  39. Ansari MA, Roberts KN, Scheff SW (2008) Oxidative stress and modification of synaptic proteins in hippocampus after traumatic brain injury. Free Radic Biol Med 45(4):443–452

    PubMed Central  CAS  PubMed  Google Scholar 

  40. Bayir H, Kochanek PM, Kagan VE (2006) Oxidative stress in immature brain after traumatic brain injury. Dev Neurosci 28(4–5):420–431

    CAS  PubMed  Google Scholar 

  41. Simonson SG, Zhang J, Canada AT, Su YF, Benveniste H, Piantadosi CA (1993) Hydrogen peroxide production by monoamine oxidase during ischemia-reperfusion in the rat brain. J Cereb Blood Flow Metab 13(1):125–134

    CAS  PubMed  Google Scholar 

  42. Balaban RS, Nemoto S, Finkel T (2005) Mitochondria, oxidants, and aging. Cell 120(4):483–495

    CAS  PubMed  Google Scholar 

  43. Jensen PK (1966) Antimycin-insensitive oxidation of succinate and reduced nicotinamide-adenine dinucleotide in electron-transport particles. I. pH dependency and hydrogen peroxide formation. Biochim Biophys Acta 122:157–166

    CAS  PubMed  Google Scholar 

  44. Chance B, Sies H, Boveris A (1979) Hydroperoxide metabolism in mammalian organs. Physiol Rev 59:527–605

    CAS  PubMed  Google Scholar 

  45. Loschen G, Flohé L, Chance B (1971) Respiratory chain linked H2O2 production in pigeon heart mitochondria. FEBS Lett 18(2):261–264

    CAS  PubMed  Google Scholar 

  46. Boveris A, Chance B (1973) The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem J 134(3):707–716

    PubMed Central  CAS  PubMed  Google Scholar 

  47. Turrens JF, Boveris A (1980) Generation of superoxide anion by the NADH dehydrogenase of bovine heart mitochondria. Biochem J 191(2):421–427

    PubMed Central  CAS  PubMed  Google Scholar 

  48. Hoek JB, Cahill A, Pastorino JG (2002) Alcohol and mitochondria: a dysfunctional relationship. Gastroenterology 122(7):2049–2063

    PubMed Central  CAS  PubMed  Google Scholar 

  49. Ho E, Galougahi KK, Liu CC, Bhindi R, Figtree GA (2013) Biological markers of oxidative stress: applications to cardiovascular research and practice. Redox Biol 1(1):483–491

    PubMed Central  CAS  PubMed  Google Scholar 

  50. Mendes Arent A, de Souza LF, Walz R, Dafre AL (2014) Perspectives on molecular biomarkers of oxidative stress and antioxidant strategies in traumatic brain injury. Biomed Res Int 2014, 723060

    PubMed Central  PubMed  Google Scholar 

  51. Morrow JD, Awad JA, Boss HJ, Blair IA, Roberts LJ 2nd (1992) Non-cyclooxygenase-derived prostanoids (F2-isoprostanes) are formed in situ on phospholipids. Proc Nat Acad Sci (USA) 89:10721–10725

    CAS  Google Scholar 

  52. Bevan RJ, Durand MF, Hickenbotham PT, Kitas GD, Patel PR, Podmore ID, Griffiths HR, Waller HL, Lunec J (2003) Validation of a novel ELISA for measurement of MDA-LDL in human plasma. Free Radic Biol Med 35:517–527

    CAS  PubMed  Google Scholar 

  53. Schopfer FJ, Baker PR, Freeman BA (2003) NO-dependent protein nitration: a cell signaling event or an oxidative inflammatory response? Trend Biochem Sci 28:646–654

    CAS  PubMed  Google Scholar 

  54. Rossi R, Dalle-Donne I, Milzani A, Giustarini D (2006) Oxidized forms of glutathione in peripheral blood as biomarkers of oxidative stress. Clin Chem 52:1406–1414

    CAS  PubMed  Google Scholar 

  55. Bursell SE, King GL (2000) The potential use of glutathionyl hemoglobin as a clinical marker of oxidative stress. Clin Chem 46:145–146

    CAS  PubMed  Google Scholar 

  56. Gavrieli Y, Sherman Y, Ben-Sasson SA (1992) Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119(3):493–501

    CAS  PubMed  Google Scholar 

  57. Khan N, Swartz H (2002) Measurements in vivo of parameters pertinent to ROS/RNS using EPR spectroscopy. Mol Cell Biochem 234–235(1–2):341–357

    PubMed  Google Scholar 

  58. Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD (2006) Blood-brain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol 1(3):223–236

    PubMed  Google Scholar 

  59. Hawkins BT, Davis TP (2005) The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 57(2):173–185

    CAS  PubMed  Google Scholar 

  60. Farkas E, Luiten PG (2001) Cerebral microvascular pathology in aging and Alzheimer’s disease. Prog Neurobiol 64(6):575–611

    CAS  PubMed  Google Scholar 

  61. de Vries HE, Kuiper J, de Boer AG, Van Berkel TJ, Breimer DD (1997) The blood-brain barrier in neuroinflammatory diseases. Pharmacol Rev 49(2):143–155

    PubMed  Google Scholar 

  62. Iłzecka J (1996) The structure and function of blood-brain barrier in ischaemic brain stroke process. Ann Univ Mariae Curie Sklodowska Med 51:123–127

    PubMed  Google Scholar 

  63. Fiala M, Liu QN, Sayre J, Pop V, Brahmandam V, Graves MC, Vinters HV (2002) Cyclooxygenase-2-positive macrophages infiltrate the Alzheimer’s disease brain and damage the blood–brain barrier. Eur J Clin Invest 32(5):360–371

    CAS  PubMed  Google Scholar 

  64. Persidsky Y, Heilman D, Haorah J, Zelivyanskaya M, Persidsky R, Weber GA, Shimokawa H, Kaibuchi K, Ikezu T (2006) Rho-mediated regulation of tight junctions during monocyte migration across the blood-brain barrier in HIV-1 encephalitis (HIVE). Blood 107(12):4770–4780

    PubMed Central  CAS  PubMed  Google Scholar 

  65. Opdenakker G, Nelissen I, Van Damme J (2003) Functional roles and therapeutic targeting of gelatinase B and chemokines in multiple sclerosis. Lancet Neurol 2(12):747–756

    CAS  PubMed  Google Scholar 

  66. Morganti-Kossmann MC, Hans VH, Lenzlinger PM, Dubs R, Ludwig E, Trentz O, Kossmann T (1999) TGF-beta is elevated in the CSF of patients with severe traumatic brain injuries and parallels blood-brain barrier function. J Neurotrauma 16(7):617–628

    CAS  PubMed  Google Scholar 

  67. Abdul Muneer PM, Alikunju S, Szlachetka AM, Haorah J (2011) Inhibitory effects of alcohol on glucose transport across the blood-brain barrier leads to neurodegeneration: preventive role of acetyl-L-carnitine. Psychopharmacology (Berl) 214(3):707–718

    CAS  Google Scholar 

  68. Abdul Muneer PM, Alikunju S, Szlachetka AM, Haorah J (2012) The mechanisms of cerebral vascular dysfunction and neuroinflammation by MMP-mediated degradation of VEGFR-2 in alcohol ingestion. Arterioscler Thromb Vasc Biol 32(5):1167–1177

    PubMed Central  CAS  PubMed  Google Scholar 

  69. Haorah J, Ramirez SH, Schall K, Smith D, Pandya R, Persidsky Y (2007) Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction. J Neurochem 101(2):566–576

    CAS  PubMed  Google Scholar 

  70. Hall ED, Vaishnav RA, Mustafa AG (2010) Antioxidant therapies for traumatic brain injury. Neurotherapeutics 7(1):51–61

    PubMed Central  CAS  PubMed  Google Scholar 

  71. Mertsch K, Blasig I, Grune T (2001) 4-Hydroxynonenal impairs the permeability of an in vitro rat blood-brain barrier. Neurosci Lett 314(3):135–138

    CAS  PubMed  Google Scholar 

  72. Smith SL, Andrus PK, Zhang JR, Hall ED (1994) Direct measurement of hydroxyl radicals, lipid peroxidation, and blood-brain barrier disruption following unilateral cortical impact head injury in the rat. J Neurotrauma 11(4):393–404

    CAS  PubMed  Google Scholar 

  73. Soares HD, Hicks RR, Smith D, McIntosh TK (1995) Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury. J Neurosci 15(12):8223–8233

    CAS  PubMed  Google Scholar 

  74. Carlos TM, Clark RS, Franicola-Higgins D, Schiding JK, Kochanek PM (1997) Expression of endothelial adhesion molecules and recruitment of neutrophils after traumatic brain injury in rats. J Leukoc Biol 61(3):279–285

    CAS  PubMed  Google Scholar 

  75. Isaksson J, Lewén A, Hillered L, Olsson Y (1997) Up-regulation of intercellular adhesion molecule 1 in cerebral microvessels after cortical contusion trauma in a rat model. Acta Neuropathol 94(1):16–20

    CAS  PubMed  Google Scholar 

  76. Marchi N, Bazarian JJ, Puvenna V, Janigro M, Ghosh C, Zhong J, Zhu T, Blackman E, Stewart D, Ellis J et al (2013) Consequences of repeated blood-brain barrier disruption in football players. PLoS One 8(3):e56805

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Kleindienst A, Ross Bullock M (2006) A critical analysis of the role of the neurotrophic protein S100B in acute brain injury. J Neurotrauma 23(8):1185–1200

    PubMed  Google Scholar 

  78. Kapural M, Krizanac-Bengez L, Barnett G, Perl J, Masaryk T, Apollo D, Rasmussen P, Mayberg MR, Janigro D (2002) Serum S-100beta as a possible marker of blood-brain barrier disruption. Brain Res 940(1–2):102–104

    CAS  PubMed  Google Scholar 

  79. Saw MM, Chamberlain J, Barr M, Morgan MP, Burnett JR, Ho KM (2014) Differential disruption of blood-brain barrier in severe traumatic brain injury. Neurocrit Care 20(2):209–216

    CAS  PubMed  Google Scholar 

  80. Bao HJ, Wang T, Zhang MY, Liu R, Dai DK, Wang YQ, Wang L, Zhang L, Gao YZ, Qin ZH et al (2012) Poloxamer-188 attenuates TBI-induced blood-brain barrier damage leading to decreased brain edema and reduced cellular death. Neurochem Res 37(12):2856–2867

    CAS  PubMed  Google Scholar 

  81. Moxon-Emre I, Schlichter LC (2011) Neutrophil depletion reduces blood-brain barrier breakdown, axon injury, and inflammation after intracerebral hemorrhage. J Neuropathol Exp Neurol 70(3):218–235

    CAS  PubMed  Google Scholar 

  82. Lopez NE, Krzyzaniak MJ, Blow C, Putnam J, Ortiz-Pomales Y, Hageny AM, Eliceiri B, Coimbra R, Bansal V (2012) Ghrelin prevents disruption of the blood-brain barrier after traumatic brain injury. J Neurotrauma 29(2):385–393

    PubMed Central  PubMed  Google Scholar 

  83. Sternlicht MD, Werb Z (2001) How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17:463–516

    PubMed Central  CAS  PubMed  Google Scholar 

  84. Nagase H, Woessner JF (1999) Matrix metalloproteinases. J Biol Chem 274(31):21491–21494

    CAS  PubMed  Google Scholar 

  85. Engel CK, Pirard B, Schimanski S, Kirsch R, Habermann J, Klingler O, Schlotte V, Weithmann KU, Wendt KU (2005) Structural basis for the highly selective inhibition of MMP-13. Chem Biol 12(2):181–189

    CAS  PubMed  Google Scholar 

  86. Visse R, Nagase H (2003) Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 92(8):827–839

    CAS  PubMed  Google Scholar 

  87. Nagase H, Visse R, Murphy G (2006) Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 69(3):562–573

    CAS  PubMed  Google Scholar 

  88. Brew K, Dinakarpandian D, Nagase H (2000) Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim Biophys Acta 1477(1–2):267–283

    CAS  PubMed  Google Scholar 

  89. Machado LS, Kozak A, Ergul A, Hess DC, Borlongan CV, Fagan SC (2006) Delayed minocycline inhibits ischemia-activated matrix metalloproteinases 2 and 9 after experimental stroke. BMC Neurosci 7:56

    PubMed Central  PubMed  Google Scholar 

  90. Kim GW, Gasche Y, Grzeschik S, Copin JC, Maier CM, Chan PH (2003) Neurodegeneration in striatum induced by the mitochondrial toxin 3-nitropropionic acid: role of matrix metalloproteinase-9 in early blood-brain barrier disruption? J Neurosci 23(25):8733–8742

    CAS  PubMed  Google Scholar 

  91. Gasche Y, Fujimura M, Morita-Fujimura Y, Copin JC, Kawase M, Massengale J, Chan PH (1999) Early appearance of activated matrix metalloproteinase-9 after focal cerebral ischemia in mice: a possible role in blood-brain barrier dysfunction. J Cereb Blood Flow Metab 19(9):1020–1028

    CAS  PubMed  Google Scholar 

  92. Galli A, Svegliati-Baroni G, Ceni E, Milani S, Ridolfi F, Salzano R, Tarocchi M, Grappone C, Pellegrini G, Benedetti A et al (2005) Oxidative stress stimulates proliferation and invasiveness of hepatic stellate cells via a MMP2-mediated mechanism. Hepatology 41(5):1074–1084

    CAS  PubMed  Google Scholar 

  93. Rosell A, Alvarez-Sabín J, Arenillas JF, Rovira A, Delgado P, Fernández-Cadenas I, Penalba A, Molina CA, Montaner J (2005) A matrix metalloproteinase protein array reveals a strong relation between MMP-9 and MMP-13 with diffusion-weighted image lesion increase in human stroke. Stroke 36(7):1415–1420

    CAS  PubMed  Google Scholar 

  94. Rosell A, Ortega-Aznar A, Alvarez-Sabin J, Fernandez-Cadenas I, Ribo M, Molina CA, Lo EH, Montaner J (2006) Increased brain expression of matrix metalloproteinase-9 after ischemic and hemorrhagic human stroke. Stroke 37:1399–1406

    CAS  PubMed  Google Scholar 

  95. Fukuda S, Fini CA, Mabuchi T, Koziol JA, Eggleston LL, del Zoppo GJ (2004) Focal cerebral ischemia induces active proteases that degrade microvascular matrix. Stroke 35(4):998–1004

    PubMed Central  CAS  PubMed  Google Scholar 

  96. Gursoy-Ozdemir Y, Qiu J, Matsuoka N, Bolay H, Bermpohl D, Jin H, Wang X, Rosenberg GA, Lo EH, Moskowitz MA (2004) Cortical spreading depression activates and upregulates MMP-9. J Clin Invest 113(10):1447–1455

    PubMed Central  CAS  PubMed  Google Scholar 

  97. Sánchez-del-Rio M, Reuter U (2004) Migraine aura: new information on underlying mechanisms. Curr Opin Neurol 17(3):289–293

    PubMed  Google Scholar 

  98. Hanumegowda UM, Copple BL, Shibuya M, Malle E, Ganey PE, Roth RA (2003) Basement membrane and matrix metalloproteinases in monocrotaline-induced liver injury. Toxicol Sci 76(1):237–246

    CAS  PubMed  Google Scholar 

  99. Reijerkerk A, Kooij G, van der Pol SM, Khazen S, Dijkstra CD, de Vries HE (2006) Diapedesis of monocytes is associated with MMP-mediated occludin disappearance in brain endothelial cells. FASEB J 20(14):2550–2552

    CAS  PubMed  Google Scholar 

  100. Grossetete M, Phelps J, Arko L, Yonas H, Rosenberg GA (2009) Elevation of matrix metalloproteinases 3 and 9 in cerebrospinal fluid and blood in patients with severe traumatic brain injury. Neurosurgery 65(4):702–708

    PubMed Central  PubMed  Google Scholar 

  101. Jia F, Pan YH, Mao Q, Liang YM, Jiang JY (2010) Matrix metalloproteinase-9 expression and protein levels after fluid percussion injury in rats: the effect of injury severity and brain temperature. J Neurotrauma 27(6):1059–1068

    PubMed  Google Scholar 

  102. Rosenberg GA (2009) Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol 8(2):205–216

    CAS  PubMed  Google Scholar 

  103. Ding JY, Kreipke CW, Schafer P, Schafer S, Speirs SL, Rafols JA (2009) Synapse loss regulated by matrix metalloproteinases in traumatic brain injury is associated with hypoxia inducible factor-1alpha expression. Brain Res 1268:125–134

    PubMed Central  CAS  PubMed  Google Scholar 

  104. Hayashi T, Kaneko Y, Yu S, Bae E, Stahl CE, Kawase T, van Loveren H, Sanberg PR, Borlongan CV (2009) Quantitative analyses of matrix metalloproteinase activity after traumatic brain injury in adult rats. Brain Res 1280:172–177

    CAS  PubMed  Google Scholar 

  105. Tejima E, Guo S, Murata Y, Arai K, Lok J, van Leyen K, Rosell A, Wang X, Lo EH (2009) Neuroprotective effects of overexpressing tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma 26(11):1935–1941

    PubMed Central  PubMed  Google Scholar 

  106. Ranaivo RH, Zunich SM, Choi N, Hodge JN, Wainwright MS (2011) Mild stretch-induced injury increases susceptibility to interleukin-1β-induced release of matrix metalloproteinase-9 from astrocytes. J Neurotrauma 28(9):1757–1766

    Google Scholar 

  107. Higashida T, Kreipke CW, Rafols JA, Peng C, Schafer S, Schafer P, Ding JY, Dornbos D, Li X, Guthikonda M et al (2011) The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg 114(1):92–101

    CAS  PubMed  Google Scholar 

  108. Tonnesen MG, Feng X, Clark RA (2000) Angiogenesis in wound healing. J Invest Dermatol Symp Proc 5:40–46

    CAS  Google Scholar 

  109. Singer AJ, Clark RA (1999) Cutaneous wound healing. N Engl J Med 341(10):738–746

    CAS  PubMed  Google Scholar 

  110. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z (1999) Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 13(1):9–22

    CAS  PubMed  Google Scholar 

  111. Baffert F, Le T, Sennino B, Thurston G, Kuo CJ, Hu-Lowe D, McDonald DM (2006) Cellular changes in normal blood capillaries undergoing regression after inhibition of VEGF signaling. Am J Physiol Heart Circ Physiol 290(2):H547–H559

    CAS  PubMed  Google Scholar 

  112. Gerber HP, Hillan KJ, Ryan AM, Kowalski J, Keller GA, Rangell L, Wright BD, Radtke F, Aguet M, Ferrara N (1999) VEGF is required for growth and survival in neonatal mice. Development 126(6):1149–1159

    CAS  PubMed  Google Scholar 

  113. Katoh O, Tauchi H, Kawaishi K, Kimura A, Satow Y (1995) Expression of the vascular endothelial growth factor (VEGF) receptor gene, KDR, in hematopoietic cells and inhibitory effect of VEGF on apoptotic cell death caused by ionizing radiation. Cancer Res 55(23):5687–5692

    CAS  PubMed  Google Scholar 

  114. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC (1995) Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 376(6535):62–66

    CAS  PubMed  Google Scholar 

  115. Haorah J, Schall K, Ramirez SH, Persidsky Y (2008) Activation of protein tyrosine kinases and matrix metalloproteinases causes blood-brain barrier injury: novel mechanism for neurodegeneration associated with alcohol abuse. Glia 56(1):78–88

    PubMed Central  PubMed  Google Scholar 

  116. Cunningham LA, Wetzel M, Rosenberg GA (2005) Multiple roles for MMPs and TIMPs in cerebral ischemia. Glia 50(4):329–339

    PubMed  Google Scholar 

  117. Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA (2007) Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab 27(4):697–709

    CAS  PubMed  Google Scholar 

  118. Valable S, Montaner J, Bellail A, Berezowski V, Brillault J, Cecchelli R, Divoux D, Mackenzie ET, Bernaudin M, Roussel S et al (2005) VEGF-induced BBB permeability is associated with an MMP-9 activity increase in cerebral ischemia: both effects decreased by Ang-1. J Cereb Blood Flow Metab 25(11):1491–1504

    CAS  PubMed  Google Scholar 

  119. Tsuji K, Aoki T, Tejima E, Arai K, Lee SR, Atochin DN, Huang PL, Wang X, Montaner J, Lo EH (2005) Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke 36(9):1954–1959

    CAS  PubMed  Google Scholar 

  120. Turk BE, Huang LL, Piro ET, Cantley LC (2001) Determination of protease cleavage site motifs using mixture-based oriented peptide libraries. Nat Biotechnol 19(7):661–667

    CAS  PubMed  Google Scholar 

  121. Tran ED, DeLano FA, Schmid-Schönbein GW (2010) Enhanced matrix metalloproteinase activity in the spontaneously hypertensive rat: VEGFR-2 cleavage, endothelial apoptosis, and capillary rarefaction. J Vasc Res 47(5):423–431

    PubMed Central  CAS  PubMed  Google Scholar 

  122. Lee C, Agoston DV (2009) Inhibition of VEGF receptor 2 increased cell death of dentate hilar neurons after traumatic brain injury. Exp Neurol 220(2):400–403

    CAS  PubMed  Google Scholar 

  123. Chodobski A, Chung I, Koźniewska E, Ivanenko T, Chang W, Harrington JF, Duncan JA, Szmydynger-Chodobska J (2003) Early neutrophilic expression of vascular endothelial growth factor after traumatic brain injury. Neuroscience 122(4):853–867

    CAS  PubMed  Google Scholar 

  124. Abid MR, Tsai JC, Spokes KC, Deshpande SS, Irani K, Aird WC (2001) Vascular endothelial growth factor induces manganese-superoxide dismutase expression in endothelial cells by a Rac1-regulated NADPH oxidase-dependent mechanism. FASEB J 15(13):2548–2550

    CAS  PubMed  Google Scholar 

  125. Thau-Zuchman O, Shohami E, Alexandrovich AG, Leker RR (2010) Vascular endothelial growth factor increases neurogenesis after traumatic brain injury. J Cereb Blood Flow Metab 30(5):1008–1016

    PubMed Central  CAS  PubMed  Google Scholar 

  126. Werner C, Engelhard K (2007) Pathophysiology of traumatic brain injury. Br J Anaesth 99(1):4–9

    CAS  PubMed  Google Scholar 

  127. Lucas SM, Rothwell NJ, Gibson RM (2006) The role of inflammation in CNS injury and disease. Br J Pharmacol 147(Suppl 1):S232–S240

    PubMed Central  CAS  PubMed  Google Scholar 

  128. Hans VH, Kossmann T, Joller H, Otto V, Morganti-Kossmann MC (1999) Interleukin-6 and its soluble receptor in serum and cerebrospinal fluid after cerebral trauma. Neuroreport 10(2):409–412

    CAS  PubMed  Google Scholar 

  129. Shohami E, Bass R, Wallach D, Yamin A, Gallily R (1996) Inhibition of tumor necrosis factor alpha (TNFalpha) activity in rat brain is associated with cerebroprotection after closed head injury. J Cereb Blood Flow Metab 16(3):378–384

    CAS  PubMed  Google Scholar 

  130. Fan L, Young PR, Barone FC, Feuerstein GZ, Smith DH, McIntosh TK (1995) Experimental brain injury induces expression of interleukin-1 beta mRNA in the rat brain. Brain Res Mol Brain Res 30:125–130

    CAS  PubMed  Google Scholar 

  131. Vecil GG, Larsen PH, Corley SM, Herx LM, Besson A, Goodyer CG, Yong VW (2000) Interleukin-1 is a key regulator of matrix metalloproteinase-9 expression in human neurons in culture and following mouse brain trauma in vivo. J Neurosci Res 61(2):212–224

    CAS  PubMed  Google Scholar 

  132. Lawrence CB, Allan SM, Rothwell NJ (1998) Interleukin-1beta and the interleukin-1 receptor antagonist act in the striatum to modify excitotoxic brain damage in the rat. Eur J Neurosci 10(3):1188–1195

    CAS  PubMed  Google Scholar 

  133. Hopkins SJ, Rothwell NJ (1995) Cytokines and the nervous system. I: expression and recognition. Trends Neurosci 18(2):83–88

    CAS  PubMed  Google Scholar 

  134. Rothwell NJ, Hopkins SJ (1995) Cytokines and the nervous system II: actions and mechanisms of action. Trends Neurosci 18(3):130–136

    CAS  PubMed  Google Scholar 

  135. Lu KT, Wang YW, Yang JT, Yang YL, Chen HI (2005) Effect of interleukin-1 on traumatic brain injury-induced damage to hippocampal neurons. J Neurotrauma 22(8):885–895

    PubMed  Google Scholar 

  136. Knoblach SM, Faden AI (2000) Cortical interleukin-1 beta elevation after traumatic brain injury in the rat: no effect of two selective antagonists on motor recovery. Neurosci Lett 289(1):5–8

    CAS  PubMed  Google Scholar 

  137. Clausen F, Hånell A, Israelsson C, Hedin J, Ebendal T, Mir AK, Gram H, Marklund N (2011) Neutralization of interleukin-1β reduces cerebral edema and tissue loss and improves late cognitive outcome following traumatic brain injury in mice. Eur J Neurosci 34(1):110–123

    PubMed  Google Scholar 

  138. Loddick SA, Rothwell NJ (1996) Neuroprotective effects of human recombinant interleukin-1 receptor antagonist in focal cerebral ischaemia in the rat. J Cereb Blood Flow Metab 16(5):932–940

    CAS  PubMed  Google Scholar 

  139. Lamkanfi M, Kanneganti TD, Van Damme P, Vanden Berghe T, Vanoverberghe I, Vandekerckhove J, Vandenabeele P, Gevaert K, Núñez G (2008) Targeted peptidecentric proteomics reveals caspase-7 as a substrate of the caspase-1 inflammasomes. Mol Cell Proteomics 7(12):2350–2363

    PubMed Central  CAS  PubMed  Google Scholar 

  140. Keller M, Rüegg A, Werner S, Beer HD (2008) Active caspase-1 is a regulator of unconventional protein secretion. Cell 132(5):818–831

    CAS  PubMed  Google Scholar 

  141. Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J (1992) A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature 356(6372):768–774

    CAS  PubMed  Google Scholar 

  142. Sifringer M, Stefovska V, Endesfelder S, Stahel PF, Genz K, Dzietko M, Ikonomidou C, Felderhoff-Mueser U (2007) Activation of caspase-1 dependent interleukins in developmental brain trauma. Neurobiol Dis 25(3):614–622

    CAS  PubMed  Google Scholar 

  143. Mejia SRO, Ona VO, Li M, Friedlander RM (2001) Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurgery 48(6):1393–1399, discussion 1399-1401

    Google Scholar 

  144. Csuka E, Morganti-Kossmann MC, Lenzlinger PM, Joller H, Trentz O, Kossmann T (1999) IL-10 levels in cerebrospinal fluid and serum of patients with severe traumatic brain injury: relationship to IL-6, TNF-alpha, TGF-beta1 and blood-brain barrier function. J Neuroimmunol 101(2):211–221

    CAS  PubMed  Google Scholar 

  145. Bell MJ, Kochanek PM, Doughty LA, Carcillo JA, Adelson PD, Clark RS, Wisniewski SR, Whalen MJ, DeKosky ST (1997) Interleukin-6 and interleukin-10 in cerebrospinal fluid after severe traumatic brain injury in children. J Neurotrauma 14(7):451–457

    CAS  PubMed  Google Scholar 

  146. Krum JM, Mani N, Rosenstein JM (2008) Roles of the endogenous VEGF receptors flt-1 and flk-1 in astroglial and vascular remodeling after brain injury. Exp Neurol 212(1):108–117

    PubMed Central  CAS  PubMed  Google Scholar 

  147. Penkowa M, Camats J, Hadberg H, Quintana A, Rojas S, Giralt M, Molinero A, Campbell IL, Hidalgo J (2003) Astrocyte-targeted expression of interleukin-6 protects the central nervous system during neuroglial degeneration induced by 6-aminonicotinamide. J Neurosci Res 73(4):481–496

    CAS  PubMed  Google Scholar 

  148. Swartz KR, Liu F, Sewell D, Schochet T, Campbell I, Sandor M, Fabry Z (2001) Interleukin-6 promotes post-traumatic healing in the central nervous system. Brain Res 896(1–2):86–95

    CAS  PubMed  Google Scholar 

  149. Ramilo O, Sáez-Llorens X, Mertsola J, Jafari H, Olsen KD, Hansen EJ, Yoshinaga M, Ohkawara S, Nariuchi H, McCracken GH (1990) Tumor necrosis factor alpha/cachectin and interleukin 1 beta initiate meningeal inflammation. J Exp Med 172(2):497–507

    CAS  PubMed  Google Scholar 

  150. Kim KS, Wass CA, Cross AS, Opal SM (1992) Modulation of blood-brain barrier permeability by tumor necrosis factor and antibody to tumor necrosis factor in the rat. Lymphokine Cytokine Res 11(6):293–298

    CAS  PubMed  Google Scholar 

  151. Ross SA, Halliday MI, Campbell GC, Byrnes DP, Rowlands BJ (1994) The presence of tumour necrosis factor in CSF and plasma after severe head injury. Br J Neurosurg 8(4):419–425

    CAS  PubMed  Google Scholar 

  152. Khuman J, Meehan WP, Zhu X, Qiu J, Hoffmann U, Zhang J, Giovannone E, Lo EH, Whalen MJ (2011) Tumor necrosis factor alpha and Fas receptor contribute to cognitive deficits independent of cell death after concussive traumatic brain injury in mice. J Cereb Blood Flow Metab 31(2):778–789

    PubMed Central  CAS  PubMed  Google Scholar 

  153. Bermpohl D, You Z, Lo EH, Kim HH, Whalen MJ (2007) TNF alpha and Fas mediate tissue damage and functional outcome after traumatic brain injury in mice. J Cereb Blood Flow Metab 27(11):1806–1818

    CAS  PubMed  Google Scholar 

  154. Logan A, Berry M (1993) Transforming growth factor-beta 1 and basic fibroblast growth factor in the injured CNS. Trends Pharmacol Sci 14(9):337–342

    CAS  PubMed  Google Scholar 

  155. Zhu Y, Roth-Eichhorn S, Braun N, Culmsee C, Rami A, Krieglstein J (2000) The expression of transforming growth factor-beta1 (TGF-beta1) in hippocampal neurons: a temporary upregulated protein level after transient forebrain ischemia in the rat. Brain Res 866(1–2):286–298

    CAS  PubMed  Google Scholar 

  156. Lehrmann E, Kiefer R, Christensen T, Toyka KV, Zimmer J, Diemer NH, Hartung HP, Finsen B (1998) Microglia and macrophages are major sources of locally produced transforming growth factor-beta1 after transient middle cerebral artery occlusion in rats. Glia 24(4):437–448

    CAS  PubMed  Google Scholar 

  157. Rimaniol AC, Lekieffre D, Serrano A, Masson A, Benavides J, Zavala F (1995) Biphasic transforming growth factor-beta production flanking the pro-inflammatory cytokine response in cerebral trauma. Neuroreport 7(1):133–136

    CAS  PubMed  Google Scholar 

  158. Morganti-Kossmann MC, Rancan M, Stahel PF, Kossmann T (2002) Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr Opin Crit Care 8(2):101–105

    PubMed  Google Scholar 

  159. Cacheaux LP, Ivens S, David Y, Lakhter AJ, Bar-Klein G, Shapira M, Heinemann U, Friedman A, Kaufer D (2009) Transcriptome profiling reveals TGF-beta signaling involvement in epileptogenesis. J Neurosci 29(28):8927–8935

    PubMed Central  CAS  PubMed  Google Scholar 

  160. Marmarou A (2003) Pathophysiology of traumatic brain edema: current concepts. Acta Neurochir Suppl 86:7–10

    CAS  PubMed  Google Scholar 

  161. Klatzo I (1987) Pathophysiological aspects of brain edema. Acta Neuropathol 72(3):236–239

    CAS  PubMed  Google Scholar 

  162. Donkin JJ, Vink R (2010) Mechanisms of cerebral edema in traumatic brain injury: therapeutic developments. Curr Opin Neurol 23(3):293–299

    CAS  PubMed  Google Scholar 

  163. Barzó P, Marmarou A, Fatouros P, Hayasaki K, Corwin F (1997) Contribution of vasogenic and cellular edema to traumatic brain swelling measured by diffusion-weighted imaging. J Neurosurg 87(6):900–907

    PubMed  Google Scholar 

  164. Unterberg AW, Stroop R, Thomale UW, Kiening KL, Päuser S, Vollmann W (1997) Characterisation of brain edema following “controlled cortical impact injury” in rats. Acta Neurochir Suppl 70:106–108

    CAS  PubMed  Google Scholar 

  165. Papadopoulos MC, Verkman AS (2008) Potential utility of aquaporin modulators for therapy of brain disorders. Prog Brain Res 170:589–601

    PubMed Central  CAS  PubMed  Google Scholar 

  166. Pasantes-Morales H, Cruz-Rangel S (2010) Brain volume regulation: osmolytes and aquaporin perspectives. Neuroscience 168(4):871–884

    CAS  PubMed  Google Scholar 

  167. Badaut J, Lasbennes F, Magistretti PJ, Regli L (2002) Aquaporins in brain: distribution, physiology, and pathophysiology. J Cereb Blood Flow Metab 22(4):367–378

    CAS  PubMed  Google Scholar 

  168. Nag S, Manias JL, Stewart DJ (2009) Pathology and new players in the pathogenesis of brain edema. Acta Neuropathol 118(2):197–217

    PubMed  Google Scholar 

  169. Tran ND, Kim S, Vincent HK, Rodriguez A, Hinton DR, Bullock MR, Young HF (2010) Aquaporin-1-mediated cerebral edema following traumatic brain injury: effects of acidosis and corticosteroid administration. J Neurosurg 112(5):1095–1104

    CAS  PubMed  Google Scholar 

  170. Aoki K, Uchihara T, Tsuchiya K, Nakamura A, Ikeda K, Wakayama Y (2003) Enhanced expression of aquaporin 4 in human brain with infarction. Acta Neuropathol 106(2):121–124

    CAS  PubMed  Google Scholar 

  171. Qiu B, Li X, Sun X, Wang Y, Jing Z, Zhang X (2014) Overexpression of aquaporin-1 aggravates hippocampal damage in mouse traumatic brain injury models. Mol Med Rep 9(3):916–922

    CAS  PubMed  Google Scholar 

  172. Ding JY, Kreipke CW, Speirs SL, Schafer P, Schafer S, Rafols JA (2009) Hypoxia-inducible factor-1alpha signaling in aquaporin upregulation after traumatic brain injury. Neurosci Lett 453(1):68–72

    PubMed Central  CAS  PubMed  Google Scholar 

  173. Manley GT, Fujimura M, Ma T, Noshita N, Filiz F, Bollen AW, Chan P, Verkman AS (2000) Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med 6(2):159–163

    CAS  PubMed  Google Scholar 

  174. Fazzina G, Amorini AM, Marmarou CR, Fukui S, Okuno K, Dunbar JG, Glisson R, Marmarou A, Kleindienst A (2010) The protein kinase C activator phorbol myristate acetate decreases brain edema by aquaporin 4 downregulation after middle cerebral artery occlusion in the rat. J Neurotrauma 27(2):453–461

    PubMed Central  PubMed  Google Scholar 

  175. Taya K, Gulsen S, Okuno K, Prieto R, Marmarou CR, Marmarou A (2008) Modulation of AQP4 expression by the selective V1a receptor antagonist, SR49059, decreases trauma-induced brain edema. Acta Neurochir Suppl 102:425–429

    PubMed  Google Scholar 

  176. Candelario-Jalil E, Yang Y, Rosenberg GA (2009) Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience 158(3):983–994

    PubMed Central  CAS  PubMed  Google Scholar 

  177. Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA, Fini ME, Lo EH (2001) Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood-brain barrier and white matter components after cerebral ischemia. J Neurosci 21(19):7724–7732

    CAS  PubMed  Google Scholar 

  178. Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, Dixon CE, Fini ME, Lo EH (2000) Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci 20(18):7037–7042

    CAS  PubMed  Google Scholar 

  179. Abbott NJ (2000) Inflammatory mediators and modulation of blood-brain barrier permeability. Cell Mol Neurobiol 20(2):131–147

    CAS  PubMed  Google Scholar 

  180. Reiter RJ (1995) Oxidative processes and antioxidative defense mechanisms in the aging brain. FASEB J 9:526–533

    CAS  PubMed  Google Scholar 

  181. Gilgun-Sherki Y, Rosenbaum Z, Melamed E, Offen D (2002) Antioxidant therapy in acute central nervous system injury: current state. Pharmacol Rev 54(2):271–284

    CAS  PubMed  Google Scholar 

  182. Faraci FM, Didion SP (2004) Vascular protection: superoxide dismutase isoforms in the vessel wall. Arterioscler Thromb Vasc Biol 24:1367–1373

    CAS  PubMed  Google Scholar 

  183. Aoyama N, Katayama Y, Kawamata T et al (2002) Effects of antioxidant, OPC-14117, on secondary cellular damage and behavioral deficits following cortical contusion in the rat. Brain Res 934(2):117–124

    CAS  PubMed  Google Scholar 

  184. Deng-Bryant Y, Singh IN, Carrico KM, Hall ED (2008) Neuroprotective effects of tempol, a catalytic scavenger of peroxynitrite-derived free radicals, in a mouse traumatic brain injury model. J Cereb Blood Flow Metab 28(6):1114–1126

    CAS  PubMed  Google Scholar 

  185. Clausen F, Marklund N, Lewen A, Hillered L (2008) The nitrone free radical scavenger NXY-059 is neuroprotective when administered after traumatic brain injury in the rat. J Neurotrauma 25(12):1449–1457

    PubMed  Google Scholar 

  186. Inci S, Ozcan OE, Kilinc K (1998) Time-level relationship for lipid peroxidation and the protective effect of α-tocopherol in experimental mild and severe brain injury. Neurosurgery 43(2):330–336

    CAS  PubMed  Google Scholar 

  187. Ikeda Y, Mochizuki Y, Nakamura Y et al (2000) Protective effect of a novel vitamin E derivative on experimental traumatic brain edema in rats—preliminary study. Acta Neurochir 76(suppl):343–345

    CAS  Google Scholar 

  188. Kabadi SV, Maher TJ (2010) Posttreatment with uridine and melatonin following traumatic brain injury reduces edema in various brain regions in rats. Ann N Y Acad Sci 1199:105–113

    CAS  PubMed  Google Scholar 

  189. Singleton RH, Yan HQ, Fellows-Mayle W, Dixon CE (2010) Resveratrol attenuates behavioral impairments and reduces cortical and hippocampal loss in a rat controlled cortical impact model of traumatic brain injury. J Neurotrauma 27(6):1091–1099

    PubMed Central  PubMed  Google Scholar 

  190. Kline AE, Massucci JL, Ma X, Zafonte RD, Dixon CE (2004) Bromocriptine reduces lipid peroxidation and enhances spatial learning and hippocampal neuron survival in a rodent model of focal brain trauma. J Neurotrauma 21(12):1712–1722

    PubMed  Google Scholar 

  191. McIntosh TK, Thomas M, Smith D, Banbury M (1992) The novel 21-aminosteroid U74006F attenuates cerebral edema and improves survival after brain injury in the rat. J Neurotrauma 9:33–46

    CAS  PubMed  Google Scholar 

  192. Chung ES, Bok E, Chung YC, Baik HH, Jin BK (2012) Cannabinoids prevent lipopolysaccharide-induced neurodegeneration in the rat substantia nigra in vivo through inhibition of microglial activation and NADPH oxidase. Brain Res 1451:110–116

    CAS  PubMed  Google Scholar 

  193. Xiao G, Wei J, Yan W, Wang W, Lu Z (2008) Improved out comes from the administration of progesterone for patients with acute severe traumatic brain injury: a randomized controlled trial. Crit Care 12(2), article R61

  194. Muehlschlegel S, Sims JR (2009) Dantrolene: mechanisms of neuroprotection and possible clinical applications in the neurointensive care unit. Neurocrit Care 10:103–115

    PubMed Central  CAS  PubMed  Google Scholar 

  195. Razmkon A, Ahmad Sadidi A, Sherafat-Kazemzadeh E, Ali Mehrafshan A, Jamali M, Malekpour B, Masoud Saghafinia M (2011) Administration of vitamin C and vitamin E in severe head injury: a randomized double-blind controlled trial. Clin Neurosurg 58:133–137

    PubMed  Google Scholar 

  196. Giacino JT, Whyte J, Bagiella E, Kalmar K, Childs N, Khademi A, Eifert B, Long D, Katz DI, Cho S, Yablon SA, Luther M, Hammond FM, Nordenbo A, Novak P, Mercer W, Maurer-Karattup P, Sherer M (2012) Placebo-controlled trial of amantadine for severe traumatic brain injury. N Engl J Med 366(9):819–826

    CAS  PubMed  Google Scholar 

  197. Zafonte R, Friedewald WT, Lee SM, Levin B, Diaz-Arrastia R, Ansel B, Eisenberg H, Timmons SD, Temkin N, Novack T, Ricker J, Merchant R, Jallo J (2009) The citicoline brain injury treatment (COBRIT) trial: design and methods. J Neurotrauma 26(12):2207–2216

    PubMed Central  PubMed  Google Scholar 

  198. Maas AIR, Murray G, Henney H 3rd, Kassem N, Legrand V et al (2006) Efficacy and safety of dexanabinol in severe traumatic brain injury: results of a phase III randomised, placebo-controlled, clinical trial. Lancet Neurol 5:38–45

    CAS  PubMed  Google Scholar 

  199. Homsi S, Federico F, Croci N, Palmier B, Plotkine M et al (2009) Minocycline effects on cerebral edema: relations with inflammatory and oxidative stress markers following traumatic brain injury in mice. Brain Res 1291:122–132

    CAS  PubMed  Google Scholar 

  200. Viscomi MT, Latini L, Florenzano F, Bernardi G, Molinari M (2008) Minocycline attenuates microglial activation but fails to mitigate degeneration in inferior olive and pontine nuclei after focal cerebellar lesion. Cerebellum 7:401–405

    CAS  PubMed  Google Scholar 

  201. Temkin NR, Anderson GD, Winn HR, Ellenbogen RG, Britz GW, Schuster J, Lucas T, Newell DW, Mansfield PN, Machamer JE, Barber J, Dikmen SS (2007) Magnesium sulfate for neuroprotection after traumatic brain injury: a randomised controlled trial. Lancet Neurol 6(1):29–38

    CAS  PubMed  Google Scholar 

  202. Vink R, Nimmo AJ (2009) Multifunctional drugs for head injury. Neurotherapeutics 6:28–42

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by NIH/NIAAA Grant R21AA020370-01A1 to J.H. and by the U.S. Army Research Office project “Army–UNL Center of Trauma Mechanics” (Contract W911NF-08-10483) to N.C.

Conflict of Interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to P. M. Abdul-Muneer or James Haorah.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abdul-Muneer, P.M., Chandra, N. & Haorah, J. Interactions of Oxidative Stress and Neurovascular Inflammation in the Pathogenesis of Traumatic Brain Injury. Mol Neurobiol 51, 966–979 (2015). https://doi.org/10.1007/s12035-014-8752-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-014-8752-3

Keywords

Navigation