Skip to main content

CNS Barriers in Neurotrauma

  • Chapter
  • First Online:
Vascular Mechanisms in CNS Trauma

Abstract

Despite significant advances in designing neuroprotective therapies, multiple clinical trials in head trauma and spinal cord injury have produced mixed results. A better understanding of the pathophysiology of central nervous system (CNS) barriers may improve clinical translation of therapies for injury of the CNS. The blood–brain barrier and blood-spinal cord barrier are formed by specialized CNS endothelial cells. These barriers play a fundamental role in restricting the entry of blood-borne factors into the CNS. However, they also function as the gateway for the delivery of neuroprotective drugs. Neurotrauma changes the properties of CNS barriers, which may significantly affect the efficacy of neuroprotective therapies. The endothelial barriers of the CNS together with the blood-cerebrospinal fluid barrier, which is predominantly formed by the epithelial cells of the choroid plexus, also restrict the influx of circulating leukocytes into the CNS. Dysfunction of these barriers resulting from injury plays a key role in the initiation and progression of inflammation that accompanies neurotrauma.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Marklund N, Bakshi A, Castelbuono DJ et al (2006) Evaluation of pharmacological treatment strategies in traumatic brain injury. Curr Pharm Des 12:1645–1680

    PubMed  CAS  Google Scholar 

  2. Schouten JW (2007) Neuroprotection in traumatic brain injury: a complex struggle against the biology of nature. Curr Opin Crit Care 13:134–142

    PubMed  Google Scholar 

  3. Tator CH (2006) Review of treatment trials in human spinal cord injury: issues, difficulties, and recommendations. Neurosurgery 59:957–982, discussion 982–957

    PubMed  Google Scholar 

  4. Hawryluk GW, Rowland J, Kwon BK et al (2008) Protection and repair of the injured spinal cord: a review of completed, ongoing, and planned clinical trials for acute spinal cord injury. Neurosurg Focus 25:E14

    PubMed  Google Scholar 

  5. Borregaard N, Sørensen OE, Theilgaard-Monch K (2007) Neutrophil granules: a library of innate immunity proteins. Trends Immunol 28:340–345

    PubMed  CAS  Google Scholar 

  6. Newby AC (2008) Metalloproteinase expression in monocytes and macrophages and its relationship to atherosclerotic plaque instability. Arterioscler Thromb Vasc Biol 28:2108–2114

    PubMed  CAS  Google Scholar 

  7. Balabanov R, Dore-Duffy P (1998) Role of the CNS microvascular pericyte in the blood–brain barrier. J Neurosci Res 53:637–644

    PubMed  CAS  Google Scholar 

  8. Mathiisen TM, Lehre KP, Danbolt NC et al (2010) The perivascular astroglial sheath provides a complete covering of the brain microvessels: an electron microscopic 3D reconstruction. Glia 58:1094–1103

    PubMed  Google Scholar 

  9. Lassmann H, Zimprich F, Vass K et al (1991) Microglial cells are a component of the perivascular glia limitans. J Neurosci Res 28:236–243

    PubMed  CAS  Google Scholar 

  10. Abbott NJ, Rönnbäck L, Hansson E (2006) Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci 7:41–53

    PubMed  CAS  Google Scholar 

  11. Winkler EA, Bell RD, Zlokovic BV (2011) Central nervous system pericytes in health and disease. Nat Neurosci 14:1398–1405

    PubMed  CAS  Google Scholar 

  12. Chodobski A, Zink BJ, Szmydynger-Chodobska J (2011) Blood–brain barrier pathophysiology in traumatic brain injury. Transl Stroke Res 2:492–516

    PubMed  CAS  Google Scholar 

  13. Daneman R, Zhou L, Kebede AA et al (2010) Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature 468:562–566

    PubMed  CAS  Google Scholar 

  14. Armulik A, Genové G, Mäe M et al (2010) Pericytes regulate the blood–brain barrier. Nature 468:557–561

    PubMed  CAS  Google Scholar 

  15. Winkler EA, Sengillo JD, Bell RD et al (2012) Blood-spinal cord barrier pericyte reductions contribute to increased capillary permeability. J Cereb Blood Flow Metab 32:1841–1852

    PubMed  CAS  Google Scholar 

  16. Ge S, Pachter JS (2006) Isolation and culture of microvascular endothelial cells from murine spinal cord. J Neuroimmunol 177:209–214

    PubMed  CAS  Google Scholar 

  17. Prockop LD, Naidu KA, Binard JE et al (1995) Selective permeability of [3H]-D-mannitol and [14C]-carboxyl-inulin across the blood–brain barrier and blood-spinal cord barrier in the rabbit. J Spinal Cord Med 18:221–226

    PubMed  CAS  Google Scholar 

  18. Campos CR, Schröter C, Wang X et al (2012) ABC transporter function and regulation at the blood-spinal cord barrier. J Cereb Blood Flow Metab 32:1559–1566

    PubMed  CAS  Google Scholar 

  19. Strazielle N, Ghersi-Egea JF (2000) Choroid plexus in the central nervous system: biology and physiopathology. J Neuropathol Exp Neurol 59:561–574

    PubMed  CAS  Google Scholar 

  20. Kratzer I, Vasiljevic A, Rey C et al (2012) Complexity and developmental changes in the expression pattern of claudins at the blood-CSF barrier. Histochem Cell Biol 138:861–879

    PubMed  CAS  Google Scholar 

  21. Amasheh S, Meiri N, Gitter AH et al (2002) Claudin-2 expression induces cation-selective channels in tight junctions of epithelial cells. J Cell Sci 115:4969–4976

    PubMed  CAS  Google Scholar 

  22. Keep RF, Jones HC (1990) A morphometric study on the development of the lateral ventricle choroid plexus, choroid plexus capillaries and ventricular ependyma in the rat. Brain Res Dev Brain Res 56:47–53

    PubMed  CAS  Google Scholar 

  23. Szmydynger-Chodobska J, Chodobski A, Johanson CE (1994) Postnatal developmental changes in blood flow to choroid plexuses and cerebral cortex of the rat. Am J Physiol 266:R1488–R1492

    PubMed  CAS  Google Scholar 

  24. Gazzin S, Strazielle N, Schmitt C et al (2008) Differential expression of the multidrug resistance-related proteins ABCB1 and ABCC1 between blood–brain interfaces. J Comp Neurol 510:497–507

    PubMed  CAS  Google Scholar 

  25. Shen H, Smith DE, Keep RF et al (2004) Immunolocalization of the proton-coupled oligopeptide transporter PEPT2 in developing rat brain. Mol Pharm 1:248–256

    PubMed  CAS  Google Scholar 

  26. Leggas M, Adachi M, Scheffer GL et al (2004) Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Mol Cell Biol 24:7612–7621

    PubMed  CAS  Google Scholar 

  27. Saatman KE, Duhaime AC, Bullock R et al (2008) Classification of traumatic brain injury for targeted therapies. J Neurotrauma 25:719–738

    PubMed  Google Scholar 

  28. Dietrich WD, Alonso O, Halley M (1994) Early microvascular and neuronal consequences of traumatic brain injury: a light and electron microscopic study in rats. J Neurotrauma 11:289–301

    PubMed  CAS  Google Scholar 

  29. Noble LJ, Wrathall JR (1989) Distribution and time course of protein extravasation in the rat spinal cord after contusive injury. Brain Res 482:57–66

    PubMed  CAS  Google Scholar 

  30. Oertel M, Kelly DF, McArthur D et al (2002) Progressive hemorrhage after head trauma: predictors and consequences of the evolving injury. J Neurosurg 96:109–116

    PubMed  Google Scholar 

  31. Kurland D, Hong C, Aarabi B et al (2012) Hemorrhagic progression of a contusion after traumatic brain injury: a review. J Neurotrauma 29:19–31

    PubMed  Google Scholar 

  32. Simard JM, Tsymbalyuk O, Ivanov A et al (2007) Endothelial sulfonylurea receptor 1-regulated NCCa-ATP channels mediate progressive hemorrhagic necrosis following spinal cord injury. J Clin Invest 117:2105–2113

    PubMed  CAS  Google Scholar 

  33. Simard JM, Kilbourne M, Tsymbalyuk O et al (2009) Key role of sulfonylurea receptor 1 in progressive secondary hemorrhage after brain contusion. J Neurotrauma 26:2257–2267

    PubMed  Google Scholar 

  34. Bașkaya MK, Rao AM, Doğan A et al (1997) The biphasic opening of the blood–brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett 226:33–36

    PubMed  Google Scholar 

  35. Kelley BJ, Lifshitz J, Povlishock JT (2007) Neuroinflammatory responses after experimental diffuse traumatic brain injury. J Neuropathol Exp Neurol 66:989–1001

    PubMed  CAS  Google Scholar 

  36. Vaz R, Sarmento A, Borges N et al (1997) Ultrastructural study of brain microvessels in patients with traumatic cerebral contusions. Acta Neurochir (Wien) 139:215–220

    CAS  Google Scholar 

  37. Preston E, Webster J, Small D (2001) Characteristics of sustained blood–brain barrier opening and tissue injury in a model for focal trauma in the rat. J Neurotrauma 18:83–92

    PubMed  CAS  Google Scholar 

  38. Tomkins O, Shelef I, Kaizerman I et al (2008) Blood–brain barrier disruption in post-traumatic epilepsy. J Neurol Neurosurg Psychiatry 79:774–777

    PubMed  CAS  Google Scholar 

  39. Whetstone WD, Hsu JY, Eisenberg M et al (2003) Blood-spinal cord barrier after spinal cord injury: relation to revascularization and wound healing. J Neurosci Res 74:227–239

    PubMed  CAS  Google Scholar 

  40. Popovich PG, Horner PJ, Mullin BB et al (1996) A quantitative spatial analysis of the blood-spinal cord barrier. I. Permeability changes after experimental spinal contusion injury. Exp Neurol 142:258–275

    PubMed  CAS  Google Scholar 

  41. Xue M, Hollenberg MD, Yong VW (2006) Combination of thrombin and matrix metalloproteinase-9 exacerbates neurotoxicity in cell culture and intracerebral hemorrhage in mice. J Neurosci 26:10281–10291

    PubMed  CAS  Google Scholar 

  42. Rao HV, Thirumangalakudi L, Desmond P et al (2007) Cyclin D1, cdk4, and Bim are involved in thrombin-induced apoptosis in cultured cortical neurons. J Neurochem 101:498–505

    PubMed  CAS  Google Scholar 

  43. Paul J, Strickland S, Melchor JP (2007) Fibrin deposition accelerates neurovascular damage and neuroinflammation in mouse models of Alzheimer’s disease. J Exp Med 204:1999–2008

    PubMed  CAS  Google Scholar 

  44. Marmarou A, Signoretti S, Fatouros PP et al (2006) Predominance of cellular edema in traumatic brain swelling in patients with severe head injuries. J Neurosurg 104:720–730

    PubMed  Google Scholar 

  45. Endo T, Suzuki S, Utsunomiya A et al (2011) Prediction of neurological recovery using apparent diffusion coefficient in cases of incomplete spinal cord injury. Neurosurgery 68:329–336

    PubMed  Google Scholar 

  46. Dore-Duffy P, Owen C, Balabanov R et al (2000) Pericyte migration from the vascular wall in response to traumatic brain injury. Microvasc Res 60:55–69

    PubMed  CAS  Google Scholar 

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

    PubMed  Google Scholar 

  48. Yang Y, Estrada EY, Thompson JF et al (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 27:697–709

    PubMed  CAS  Google Scholar 

  49. Rosenberg GA, Yang Y (2007) Vasogenic edema due to tight junction disruption by matrix metalloproteinases in cerebral ischemia. Neurosurg Focus 22:E4

    PubMed  Google Scholar 

  50. Rosenberg GA, Cunningham LA, Wallace J et al (2001) Immunohistochemistry of matrix metalloproteinases in reperfusion injury to rat brain: activation of MMP-9 linked to stromelysin-1 and microglia in cell cultures. Brain Res 893:104–112

    PubMed  CAS  Google Scholar 

  51. Noble LJ, Donovan F, Igarashi T et al (2002) Matrix metalloproteinases limit functional recovery after spinal cord injury by modulation of early vascular events. J Neurosci 22:7526–7535

    PubMed  CAS  Google Scholar 

  52. Wells JE, Rice TK, Nuttall RK et al (2003) An adverse role for matrix metalloproteinase 12 after spinal cord injury in mice. J Neurosci 23:10107–10115

    PubMed  CAS  Google Scholar 

  53. Takata F, Dohgu S, Matsumoto J et al (2011) Brain pericytes among cells constituting the blood–brain barrier are highly sensitive to tumor necrosis factor-α, releasing matrix metalloproteinase-9 and migrating in vitro. J Neuroinflammation 8:106

    PubMed  CAS  Google Scholar 

  54. Reijerkerk A, Kooij G, van der Pol SM et al (2006) Diapedesis of monocytes is associated with MMP-mediated occludin disappearance in brain endothelial cells. FASEB J 20:2550–2552

    PubMed  CAS  Google Scholar 

  55. Truettner JS, Alonso OF, Dalton Dietrich W (2005) Influence of therapeutic hypothermia on matrix metalloproteinase activity after traumatic brain injury in rats. J Cereb Blood Flow Metab 25:1505–1516

    PubMed  CAS  Google Scholar 

  56. Goussev S, Hsu JY, Lin Y et al (2003) Differential temporal expression of matrix metalloproteinases after spinal cord injury: relationship to revascularization and wound healing. J Neurosurg 99:188–197

    PubMed  CAS  Google Scholar 

  57. Wang X, Jung J, Asahi M et al (2000) Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci 20:7037–7042

    PubMed  CAS  Google Scholar 

  58. Asahi M, Asahi K, Jung JC et al (2000) Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab 20:1681–1689

    PubMed  CAS  Google Scholar 

  59. Hsu JY, McKeon R, Goussev S et al (2006) Matrix metalloproteinase-2 facilitates wound healing events that promote functional recovery after spinal cord injury. J Neurosci 26:9841–9850

    PubMed  CAS  Google Scholar 

  60. Zhao BQ, Wang S, Kim HY et al (2006) Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 12:441–445

    PubMed  CAS  Google Scholar 

  61. Livingston DH, Lavery RF, Mosenthal AC et al (2005) Recovery at one year following isolated traumatic brain injury: a Western Trauma Association prospective multicenter trial. J Trauma 59:1298–1304, discussion 1304

    PubMed  Google Scholar 

  62. Rapoport MJ, Herrmann N, Shammi P et al (2006) Outcome after traumatic brain injury sustained in older adulthood: a one-year longitudinal study. Am J Geriatr Psychiatry 14:456–465

    PubMed  Google Scholar 

  63. Hamm RJ, Jenkins LW, Lyeth BG et al (1991) The effect of age on outcome following traumatic brain injury in rats. J Neurosurg 75:916–921

    PubMed  CAS  Google Scholar 

  64. Onyszchuk G, He YY, Berman NE et al (2008) Detrimental effects of aging on outcome from traumatic brain injury: a behavioral, magnetic resonance imaging, and histological study in mice. J Neurotrauma 25:153–171

    PubMed  Google Scholar 

  65. Kumar A, Stoica BA, Sabirzhanov B et al (2013) Traumatic brain injury in aged animals increases lesion size and chronically alters microglial/macrophage classical and alternative activation states. Neurobiol Aging 34:1397–1411

    PubMed  CAS  Google Scholar 

  66. Lee P, Kim J, Williams R et al (2012) Effects of aging on blood brain barrier and matrix metalloproteases following controlled cortical impact in mice. Exp Neurol 234:50–61

    PubMed  CAS  Google Scholar 

  67. Kidwell CS, Latour L, Saver JL et al (2008) Thrombolytic toxicity: blood brain barrier disruption in human ischemic stroke. Cerebrovasc Dis 25:338–343

    PubMed  CAS  Google Scholar 

  68. Yepes M, Roussel BD, Ali C et al (2009) Tissue-type plasminogen activator in the ischemic brain: more than a thrombolytic. Trends Neurosci 32:48–55

    PubMed  CAS  Google Scholar 

  69. Samson AL, Medcalf RL (2006) Tissue-type plasminogen activator: a multifaceted modulator of neurotransmission and synaptic plasticity. Neuron 50:673–678

    PubMed  CAS  Google Scholar 

  70. Benchenane K, Berezowski V, Ali C et al (2005) Tissue-type plasminogen activator crosses the intact blood–brain barrier by low-density lipoprotein receptor-related protein-mediated transcytosis. Circulation 111:2241–2249

    PubMed  CAS  Google Scholar 

  71. Zlokovic BV, Wang L, Sun N et al (1995) Expression of tissue plasminogen activator in cerebral capillaries: possible fibrinolytic function of the blood–brain barrier. Neurosurgery 37:955–961

    PubMed  CAS  Google Scholar 

  72. Kim JA, Tran ND, Li Z et al (2006) Brain endothelial hemostasis regulation by pericytes. J Cereb Blood Flow Metab 26:209–217

    PubMed  Google Scholar 

  73. Tran ND, Schreiber SS, Fisher M (1998) Astrocyte regulation of endothelial tissue plasminogen activator in a blood–brain barrier model. J Cereb Blood Flow Metab 18:1316–1324

    PubMed  CAS  Google Scholar 

  74. Faber-Elman A, Miskin R, Schwartz M (1995) Components of the plasminogen activator system in astrocytes are modulated by tumor necrosis factor-alpha and interleukin-1 beta through similar signal transduction pathways. J Neurochem 65:1524–1535

    PubMed  CAS  Google Scholar 

  75. Cassé F, Bardou I, Danglot L et al (2012) Glutamate controls tPA recycling by astrocytes, which in turn influences glutamatergic signals. J Neurosci 32:5186–5199

    PubMed  Google Scholar 

  76. Tsirka SE, Rogove AD, Bugge TH et al (1997) An extracellular proteolytic cascade promotes neuronal degeneration in the mouse hippocampus. J Neurosci 17:543–552

    PubMed  CAS  Google Scholar 

  77. Yepes M, Sandkvist M, Moore EG et al (2003) Tissue-type plasminogen activator induces opening of the blood–brain barrier via the LDL receptor-related protein. J Clin Invest 112:1533–1540

    PubMed  CAS  Google Scholar 

  78. Su EJ, Fredriksson L, Geyer M et al (2008) Activation of PDGF-CC by tissue plasminogen activator impairs blood–brain barrier integrity during ischemic stroke. Nat Med 14:731–737

    PubMed  CAS  Google Scholar 

  79. Polavarapu R, Gongora MC, Yi H et al (2007) Tissue-type plasminogen activator-mediated shedding of astrocytic low-density lipoprotein receptor-related protein increases the permeability of the neurovascular unit. Blood 109:3270–3278

    PubMed  CAS  Google Scholar 

  80. Sashindranath M, Sales E, Daglas M et al (2012) The tissue-type plasminogen activator-plasminogen activator inhibitor 1 complex promotes neurovascular injury in brain trauma: evidence from mice and humans. Brain 135:3251–3264

    PubMed  Google Scholar 

  81. Lee SR, Guo SZ, Scannevin RH et al (2007) Induction of matrix metalloproteinase, cytokines and chemokines in rat cortical astrocytes exposed to plasminogen activators. Neurosci Lett 417:1–5

    PubMed  CAS  Google Scholar 

  82. Wang X, Lee SR, Arai K et al (2003) Lipoprotein receptor-mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat Med 9:1313–1317

    PubMed  CAS  Google Scholar 

  83. Suzuki Y, Nagai N, Yamakawa K et al (2009) Tissue-type plasminogen activator (t-PA) induces stromelysin-1 (MMP-3) in endothelial cells through activation of lipoprotein receptor-related protein. Blood 114:3352–3358

    PubMed  CAS  Google Scholar 

  84. Hu K, Yang J, Tanaka S et al (2006) Tissue-type plasminogen activator acts as a cytokine that triggers intracellular signal transduction and induces matrix metalloproteinase-9 gene expression. J Biol Chem 281:2120–2127

    PubMed  CAS  Google Scholar 

  85. Fredriksson L, Li H, Fieber C et al (2004) Tissue plasminogen activator is a potent activator of PDGF-CC. EMBO J 23:3793–3802

    PubMed  CAS  Google Scholar 

  86. Li X, Ponten A, Aase K et al (2000) PDGF-C is a new protease-activated ligand for the PDGF alpha-receptor. Nat Cell Biol 2:302–309

    PubMed  CAS  Google Scholar 

  87. Siao CJ, Tsirka SE (2002) Tissue plasminogen activator mediates microglial activation via its finger domain through annexin II. J Neurosci 22:3352–3358

    PubMed  CAS  Google Scholar 

  88. Reijerkerk A, Kooij G, van der Pol SM et al (2008) Tissue-type plasminogen activator is a regulator of monocyte diapedesis through the brain endothelial barrier. J Immunol 181:3567–3574

    PubMed  CAS  Google Scholar 

  89. Sheehan JJ, Zhou C, Gravanis I et al (2007) Proteolytic activation of monocyte chemoattractant protein-1 by plasmin underlies excitotoxic neurodegeneration in mice. J Neurosci 27:1738–1745

    PubMed  CAS  Google Scholar 

  90. Stamatovic SM, Keep RF, Kunkel SL et al (2003) Potential role of MCP-1 in endothelial cell tight junction ‘opening’: signaling via Rho and Rho kinase. J Cell Sci 116:4615–4628

    PubMed  CAS  Google Scholar 

  91. Yao Y, Tsirka SE (2011) Truncation of monocyte chemoattractant protein 1 by plasmin promotes blood–brain barrier disruption. J Cell Sci 124:1486–1495

    PubMed  CAS  Google Scholar 

  92. Sashindranath M, Samson AL, Downes CE et al (2011) Compartment- and context-specific changes in tissue-type plasminogen activator (tPA) activity following brain injury and pharmacological stimulation. Lab Invest 91:1079–1091

    PubMed  CAS  Google Scholar 

  93. Bukhari N, Torres L, Robinson JK et al (2011) Axonal regrowth after spinal cord injury via chondroitinase and the tissue plasminogen activator (tPA)/plasmin system. J Neurosci 31:14931–14943

    PubMed  CAS  Google Scholar 

  94. Abe Y, Nakamura H, Yoshino O et al (2003) Decreased neural damage after spinal cord injury in tPA-deficient mice. J Neurotrauma 20:43–57

    PubMed  Google Scholar 

  95. Mori T, Wang X, Kline AE et al (2001) Reduced cortical injury and edema in tissue plasminogen activator knockout mice after brain trauma. Neuroreport 12:4117–4120

    PubMed  CAS  Google Scholar 

  96. Srivastava RA, Bhasin N, Srivastava N (1996) Apolipoprotein E gene expression in various tissues of mouse and regulation by estrogen. Biochem Mol Biol Int 38:91–101

    PubMed  CAS  Google Scholar 

  97. Iwata A, Browne KD, Chen XH et al (2005) Traumatic brain injury induces biphasic upregulation of ApoE and ApoJ protein in rats. J Neurosci Res 82:103–114

    PubMed  CAS  Google Scholar 

  98. Eisenberg DT, Kuzawa CW, Hayes MG (2010) Worldwide allele frequencies of the human apolipoprotein E gene: climate, local adaptations, and evolutionary history. Am J Phys Anthropol 143:100–111

    PubMed  Google Scholar 

  99. Müller K, Ingebrigtsen T, Wilsgaard T et al (2009) Prediction of time trends in recovery of cognitive function after mild head injury. Neurosurgery 64:698–704, discussion 704

    PubMed  Google Scholar 

  100. Teasdale GM, Nicoll JA, Murray G et al (1997) Association of apolipoprotein E polymorphism with outcome after head injury. Lancet 350:1069–1071

    PubMed  CAS  Google Scholar 

  101. Alexander S, Kerr ME, Kim Y et al (2007) Apolipoprotein E4 allele presence and functional outcome after severe traumatic brain injury. J Neurotrauma 24:790–797

    PubMed  Google Scholar 

  102. Jha A, Lammertse DP, Coll JR et al (2008) Apolipoprotein E ε4 allele and outcomes of traumatic spinal cord injury. J Spinal Cord Med 31:171–176

    PubMed  Google Scholar 

  103. Sun C, Ji G, Liu Q et al (2011) Apolipoprotein E epsilon 4 allele and outcomes of traumatic spinal cord injury in a Chinese Han population. Mol Biol Rep 38:4793–4796

    PubMed  CAS  Google Scholar 

  104. Moran LM, Taylor HG, Ganesalingam K et al (2009) Apolipoprotein E4 as a predictor of outcomes in pediatric mild traumatic brain injury. J Neurotrauma 26:1489–1495

    PubMed  Google Scholar 

  105. Chamelian L, Reis M, Feinstein A (2004) Six-month recovery from mild to moderate traumatic brain injury: the role of APOE-ε4 allele. Brain 127:2621–2628

    PubMed  Google Scholar 

  106. Teasdale GM, Murray GD, Nicoll JA (2005) The association between APOE ε4, age and outcome after head injury: a prospective cohort study. Brain 128:2556–2561

    PubMed  CAS  Google Scholar 

  107. Corder EH, Saunders AM, Strittmatter WJ et al (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261:921–923

    PubMed  CAS  Google Scholar 

  108. Roberts GW, Gentleman SM, Lynch A et al (1994) β amyloid protein deposition in the brain after severe head injury: implications for the pathogenesis of Alzheimer’s disease. J Neurol Neurosurg Psychiatry 57:419–425

    PubMed  CAS  Google Scholar 

  109. Ikonomovic MD, Uryu K, Abrahamson EE et al (2004) Alzheimer’s pathology in human temporal cortex surgically excised after severe brain injury. Exp Neurol 190:192–203

    PubMed  CAS  Google Scholar 

  110. Hartman RE, Laurer H, Longhi L et al (2002) Apolipoprotein E4 influences amyloid deposition but not cell loss after traumatic brain injury in a mouse model of Alzheimer’s disease. J Neurosci 22:10083–10087

    PubMed  CAS  Google Scholar 

  111. Seitz A, Kragol M, Aglow E et al (2003) Apolipoprotein E expression after spinal cord injury in the mouse. J Neurosci Res 71:417–426

    PubMed  CAS  Google Scholar 

  112. Chen Y, Lomnitski L, Michaelson DM et al (1997) Motor and cognitive deficits in apolipoprotein E-deficient mice after closed head injury. Neuroscience 80:1255–1262

    PubMed  CAS  Google Scholar 

  113. Lynch JR, Wang H, Mace B et al (2005) A novel therapeutic derived from apolipoprotein E reduces brain inflammation and improves outcome after closed head injury. Exp Neurol 192:109–116

    PubMed  CAS  Google Scholar 

  114. Hoane MR, Kaufman N, Vitek MP et al (2009) COG1410 improves cognitive performance and reduces cortical neuronal loss in the traumatically injured brain. J Neurotrauma 26:121–129

    PubMed  Google Scholar 

  115. Kaufman NA, Beare JE, Tan AA et al (2010) COG1410, an apolipoprotein E-based peptide, improves cognitive performance and reduces cortical loss following moderate fluid percussion injury in the rat. Behav Brain Res 214:395–401

    PubMed  CAS  Google Scholar 

  116. Lynch JR, Pineda JA, Morgan D et al (2002) Apolipoprotein E affects the central nervous system response to injury and the development of cerebral edema. Ann Neurol 51:113–117

    PubMed  CAS  Google Scholar 

  117. Lynch JR, Tang W, Wang H et al (2003) APOE genotype and an ApoE-mimetic peptide modify the systemic and central nervous system inflammatory response. J Biol Chem 278:48529–48533

    PubMed  CAS  Google Scholar 

  118. Fullerton SM, Shirman GA, Strittmatter WJ et al (2001) Impairment of the blood-nerve and blood–brain barriers in apolipoprotein E knockout mice. Exp Neurol 169:13–22

    PubMed  CAS  Google Scholar 

  119. Hafezi-Moghadam A, Thomas KL, Wagner DD (2007) ApoE deficiency leads to a progressive age-dependent blood–brain barrier leakage. Am J Physiol Cell Physiol 292:C1256–C1262

    PubMed  CAS  Google Scholar 

  120. Nishitsuji K, Hosono T, Nakamura T et al (2011) Apolipoprotein E regulates the integrity of tight junctions in an isoform-dependent manner in an in vitro blood–brain barrier model. J Biol Chem 286:17536–17542

    PubMed  CAS  Google Scholar 

  121. Suzuki T, Elias BC, Seth A et al (2009) PKCη regulates occludin phosphorylation and epithelial tight junction integrity. Proc Natl Acad Sci U S A 106:61–66

    PubMed  CAS  Google Scholar 

  122. Bell RD, Winkler EA, Singh I et al (2012) Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 485:512–516

    PubMed  CAS  Google Scholar 

  123. Handschumacher RE, Harding MW, Rice J et al (1984) Cyclophilin: a specific cytosolic binding protein for cyclosporin A. Science 226:544–547

    PubMed  CAS  Google Scholar 

  124. Jin ZG, Lungu AO, Xie L et al (2004) Cyclophilin A is a proinflammatory cytokine that activates endothelial cells. Arterioscler Thromb Vasc Biol 24:1186–1191

    PubMed  CAS  Google Scholar 

  125. Heine SJ, Olive D, Gao JL et al (2011) Cyclophilin A cooperates with MIP-2 to augment neutrophil migration. J Inflamm Res 4:93–104

    PubMed  CAS  Google Scholar 

  126. Pineau I, Lacroix S (2009) Endogenous signals initiating inflammation in the injured nervous system. Glia 57:351–361

    PubMed  Google Scholar 

  127. Au AK, Aneja RK, Bell MJ et al (2012) Cerebrospinal fluid levels of high-mobility group box 1 and cytochrome C predict outcome after pediatric traumatic brain injury. J Neurotrauma 29:2013–2021

    PubMed  Google Scholar 

  128. Wang H, Bloom O, Zhang M et al (1999) HMG-1 as a late mediator of endotoxin lethality in mice. Science 285:248–251

    PubMed  CAS  Google Scholar 

  129. Nagyőszi P, Wilhelm I, Farkas AE et al (2010) Expression and regulation of toll-like receptors in cerebral endothelial cells. Neurochem Int 57:556–564

    PubMed  Google Scholar 

  130. Farina C, Aloisi F, Meinl E (2007) Astrocytes are active players in cerebral innate immunity. Trends Immunol 28:138–145

    PubMed  CAS  Google Scholar 

  131. Sumi N, Nishioku T, Takata F et al (2010) Lipopolysaccharide-activated microglia induce dysfunction of the blood–brain barrier in rat microvascular endothelial cells co-cultured with microglia. Cell Mol Neurobiol 30:247–253

    PubMed  CAS  Google Scholar 

  132. Kilic U, Kilic E, Matter CM et al (2008) TLR-4 deficiency protects against focal cerebral ischemia and axotomy-induced neurodegeneration. Neurobiol Dis 31:33–40

    PubMed  CAS  Google Scholar 

  133. Hyakkoku K, Hamanaka J, Tsuruma K et al (2010) Toll-like receptor 4 (TLR4), but not TLR3 or TLR9, knock-out mice have neuroprotective effects against focal cerebral ischemia. Neuroscience 171:258–267

    PubMed  CAS  Google Scholar 

  134. Caso JR, Pradillo JM, Hurtado O et al (2007) Toll-like receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation 115:1599–1608

    PubMed  CAS  Google Scholar 

  135. Yu ZQ, Zha JH (2012) Genetic ablation of toll-like receptor 2 reduces secondary brain injury caused by cortical contusion in mice. Ann Clin Lab Sci 42:26–33

    PubMed  Google Scholar 

  136. Ziegler G, Harhausen D, Schepers C et al (2007) TLR2 has a detrimental role in mouse transient focal cerebral ischemia. Biochem Biophys Res Commun 359:574–579

    PubMed  CAS  Google Scholar 

  137. Kigerl KA, Lai W, Rivest S et al (2007) Toll-like receptor (TLR)-2 and TLR-4 regulate inflammation, gliosis, and myelin sparing after spinal cord injury. J Neurochem 102:37–50

    PubMed  CAS  Google Scholar 

  138. Stammers AT, Liu J, Kwon BK (2012) Expression of inflammatory cytokines following acute spinal cord injury in a rodent model. J Neurosci Res 90:782–790

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  140. Royo NC, Wahl F, Stutzmann JM (1999) Kinetics of polymorphonuclear neutrophil infiltration after a traumatic brain injury in rat. Neuroreport 10:1363–1367

    PubMed  CAS  Google Scholar 

  141. Beck KD, Nguyen HX, Galvan MD et al (2010) Quantitative analysis of cellular inflammation after traumatic spinal cord injury: evidence for a multiphasic inflammatory response in the acute to chronic environment. Brain 133:433–447

    PubMed  Google Scholar 

  142. Szmydynger-Chodobska J, Fox LM, Lynch KM et al (2010) Vasopressin amplifies the production of proinflammatory mediators in traumatic brain injury. J Neurotrauma 27:1449–1461

    PubMed  Google Scholar 

  143. Semple BD, Bye N, Ziebell JM et al (2010) Deficiency of the chemokine receptor CXCR2 attenuates neutrophil infiltration and cortical damage following closed head injury. Neurobiol Dis 40:394–403

    PubMed  CAS  Google Scholar 

  144. Kenne E, Erlandsson A, Lindbom L et al (2012) Neutrophil depletion reduces edema formation and tissue loss following traumatic brain injury in mice. J Neuroinflammation 9:17

    PubMed  Google Scholar 

  145. Semple BD, Bye N, Rancan M et al (2010) Role of CCL2 (MCP-1) in traumatic brain injury (TBI): evidence from severe TBI patients and CCL2−/− mice. J Cereb Blood Flow Metab 30:769–782

    PubMed  Google Scholar 

  146. Utagawa A, Bramlett HM, Daniels L et al (2008) Transient blockage of the CD11d/CD18 integrin reduces contusion volume and macrophage infiltration after traumatic brain injury in rats. Brain Res 1207:155–163

    PubMed  CAS  Google Scholar 

  147. Bao F, Shultz SR, Hepburn JD et al (2012) A CD11d monoclonal antibody treatment reduces tissue injury and improves neurological outcome after fluid percussion brain injury in rats. J Neurotrauma 29:2375–2392

    PubMed  Google Scholar 

  148. Popovich PG, Guan Z, Wei P et al (1999) Depletion of hematogenous macrophages promotes partial hindlimb recovery and neuroanatomical repair after experimental spinal cord injury. Exp Neurol 158:351–365

    PubMed  CAS  Google Scholar 

  149. Lee SM, Rosen S, Weinstein P et al (2011) Prevention of both neutrophil and monocyte recruitment promotes recovery after spinal cord injury. J Neurotrauma 28:1893–1907

    PubMed  Google Scholar 

  150. Gris D, Marsh DR, Oatway MA et al (2004) Transient blockade of the CD11d/CD18 integrin reduces secondary damage after spinal cord injury, improving sensory, autonomic, and motor function. J Neurosci 24:4043–4051

    PubMed  CAS  Google Scholar 

  151. Geremia NM, Bao F, Rosenzweig TE et al (2012) CD11d antibody treatment improves recovery in spinal cord-injured mice. J Neurotrauma 29:539–550

    PubMed  Google Scholar 

  152. Stirling DP, Liu S, Kubes P et al (2009) Depletion of Ly6G/Gr-1 leukocytes after spinal cord injury in mice alters wound healing and worsens neurological outcome. J Neurosci 29:753–764

    PubMed  CAS  Google Scholar 

  153. Martinez FO, Sica A, Mantovani A et al (2008) Macrophage activation and polarization. Front Biosci 13:453–461

    PubMed  CAS  Google Scholar 

  154. Kigerl KA, Gensel JC, Ankeny DP et al (2009) Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci 29:13435–13444

    PubMed  CAS  Google Scholar 

  155. Weckbach S, Neher M, Losacco JT et al (2012) Challenging the role of adaptive immunity in neurotrauma: Rag1 −/− mice lacking mature B and T cells do not show neuroprotection after closed head injury. J Neurotrauma 29:1233–1242

    PubMed  Google Scholar 

  156. Ankeny DP, Popovich PG (2009) Mechanisms and implications of adaptive immune responses after traumatic spinal cord injury. Neuroscience 158:1112–1121

    PubMed  CAS  Google Scholar 

  157. Jones TB, Basso DM, Sodhi A et al (2002) Pathological CNS autoimmune disease triggered by traumatic spinal cord injury: implications for autoimmune vaccine therapy. J Neurosci 22:2690–2700

    PubMed  CAS  Google Scholar 

  158. Hauben E, Nevo U, Yoles E et al (2000) Autoimmune T cells as potential neuroprotective therapy for spinal cord injury. Lancet 355:286–287

    PubMed  CAS  Google Scholar 

  159. Hauben E, Butovsky O, Nevo U et al (2000) Passive or active immunization with myelin basic protein promotes recovery from spinal cord contusion. J Neurosci 20:6421–6430

    PubMed  CAS  Google Scholar 

  160. Szmydynger-Chodobska J, Strazielle N, Zink BJ et al (2009) The role of the choroid plexus in neutrophil invasion after traumatic brain injury. J Cereb Blood Flow Metab 29:1503–1516

    PubMed  CAS  Google Scholar 

  161. Szmydynger-Chodobska J, Strazielle N, Gandy JR et al (2012) Posttraumatic invasion of monocytes across the blood-cerebrospinal fluid barrier. J Cereb Blood Flow Metab 32:93–104

    PubMed  CAS  Google Scholar 

  162. McCormick BA, Hofman PM, Kim J et al (1995) Surface attachment of Salmonella typhimurium to intestinal epithelia imprints the subepithelial matrix with gradients chemotactic for neutrophils. J Cell Biol 131:1599–1608

    PubMed  CAS  Google Scholar 

  163. McCormick BA, Parkos CA, Colgan SP et al (1998) Apical secretion of a pathogen-elicited epithelial chemoattractant activity in response to surface colonization of intestinal epithelia by Salmonella typhimurium. J Immunol 160:455–466

    PubMed  CAS  Google Scholar 

  164. Dulin JN, Moore ML, Grill RJ (2013) The dual cyclooxygenase/5-lipoxygenase inhibitor licofelone attenuates p-glycoprotein-mediated drug resistance in the injured spinal cord. J Neurotrauma 30:211–226

    PubMed  Google Scholar 

  165. Spudich A, Kilic E, Xing H et al (2006) Inhibition of multidrug resistance transporter-1 facilitates neuroprotective therapies after focal cerebral ischemia. Nat Neurosci 9:487–488

    PubMed  CAS  Google Scholar 

  166. Pop V, Sorensen DW, Kamper JE et al (2013) Early brain injury alters the blood–brain barrier phenotype in parallel with β-amyloid and cognitive changes in adulthood. J Cereb Blood Flow Metab 33:205–214

    PubMed  CAS  Google Scholar 

  167. Miller DS, Bauer B, Hartz AM (2008) Modulation of P-glycoprotein at the blood–brain barrier: opportunities to improve central nervous system pharmacotherapy. Pharmacol Rev 60:196–209

    PubMed  CAS  Google Scholar 

  168. Potschka H (2010) Targeting regulation of ABC efflux transporters in brain diseases: a novel therapeutic approach. Pharmacol Ther 125:118–127

    PubMed  CAS  Google Scholar 

  169. Simard JM, Tarasov KV, Gerzanich V (2007) Non-selective cation channels, transient receptor potential channels and ischemic stroke. Biochim Biophys Acta 1772:947–957

    PubMed  CAS  Google Scholar 

  170. Woo SK, Kwon MS, Ivanov A et al (2013) The sulfonylurea receptor 1 (Sur1)-transient receptor potential melastatin 4 (Trpm4) channel. J Biol Chem 288:3655–3667

    PubMed  CAS  Google Scholar 

  171. Sala-Rabanal M, Wang S, Nichols CG (2012) On potential interactions between non-selective cation channel TRPM4 and sulfonylurea receptor SUR1. J Biol Chem 287:8746–8756

    PubMed  CAS  Google Scholar 

  172. Simard JM, Chen M, Tarasov KV et al (2006) Newly expressed SUR1-regulated NCCa-ATP channel mediates cerebral edema after ischemic stroke. Nat Med 12:433–440

    PubMed  CAS  Google Scholar 

  173. Simard JM, Geng Z, Woo SK et al (2009) Glibenclamide reduces inflammation, vasogenic edema, and caspase-3 activation after subarachnoid hemorrhage. J Cereb Blood Flow Metab 29:317–330

    PubMed  CAS  Google Scholar 

  174. Gerzanich V, Woo SK, Vennekens R et al (2009) De novo expression of Trpm4 initiates secondary hemorrhage in spinal cord injury. Nat Med 15:185–191

    PubMed  CAS  Google Scholar 

  175. Simard JM, Yurovsky V, Tsymbalyuk N et al (2009) Protective effect of delayed treatment with low-dose glibenclamide in three models of ischemic stroke. Stroke 40:604–609

    PubMed  CAS  Google Scholar 

  176. Patel AD, Gerzanich V, Geng Z et al (2010) Glibenclamide reduces hippocampal injury and preserves rapid spatial learning in a model of traumatic brain injury. J Neuropathol Exp Neurol 69:1177–1190

    PubMed  CAS  Google Scholar 

  177. Ransohoff RM (2009) In the beginning. Nature 462:41–42

    PubMed  CAS  Google Scholar 

  178. Guo S, Kim WJ, Lok J et al (2008) Neuroprotection via matrix-trophic coupling between cerebral endothelial cells and neurons. Proc Natl Acad Sci U S A 105:7582–7587

    PubMed  CAS  Google Scholar 

  179. Sawada N, Kim HH, Moskowitz MA et al (2009) Rac1 is a critical mediator of endothelium-derived neurotrophic activity. Sci Signal 2:ra10

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Adam Chodobski .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2014 Springer Science+Business Media New York

About this chapter

Cite this chapter

Chodobski, A., Zink, B.J., Szmydynger-Chodobska, J. (2014). CNS Barriers in Neurotrauma. In: Lo, E., Lok, J., Ning, M., Whalen, M. (eds) Vascular Mechanisms in CNS Trauma. Springer Series in Translational Stroke Research, vol 5. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-8690-9_1

Download citation

  • DOI: https://doi.org/10.1007/978-1-4614-8690-9_1

  • Published:

  • Publisher Name: Springer, New York, NY

  • Print ISBN: 978-1-4614-8689-3

  • Online ISBN: 978-1-4614-8690-9

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics