Skip to main content
Erschienen in: Translational Stroke Research 3/2014

01.06.2014 | Original Article

Vascular Neural Network Phenotypic Transformation After Traumatic Injury: Potential Role in Long-Term Sequelae

verfasst von: J. Badaut, G. J. Bix

Erschienen in: Translational Stroke Research | Ausgabe 3/2014

Einloggen, um Zugang zu erhalten

Abstract

The classical neurovascular unit (NVU), composed primarily of endothelium, astrocytes, and neurons, could be expanded to include smooth muscle and perivascular nerves present in both the up- and downstream feeding blood vessels (arteries and veins). The extended NVU, which can be defined as the vascular neural network (VNN), may represent a new physiological unit to consider for therapeutic development in stroke, traumatic brain injury, and other brain disorders (Zhang et al., Nat Rev Neurol 8(12):711–716, 2012). This review is focused on traumatic brain injury and resultant post-traumatic changes in cerebral blood flow, smooth muscle cells, matrix, blood–brain barrier structures and function, and the association of these changes with cognitive outcomes as described in clinical and experimental reports. We suggest that studies characterizing TBI outcomes should increase their focus on changes to the VNN, as this may yield meaningful therapeutic targets to resolve posttraumatic dysfunction.
Literatur
2.
Zurück zum Zitat Coronado VG, Xu L, Basavaraju SV, McGuire LC, Wald MM, Faul MD, et al. Surveillance for traumatic brain injury-related deaths—United States, 1997–2007. MMWR Surveill Summ. 2011;60(5):1–32.PubMed Coronado VG, Xu L, Basavaraju SV, McGuire LC, Wald MM, Faul MD, et al. Surveillance for traumatic brain injury-related deaths—United States, 1997–2007. MMWR Surveill Summ. 2011;60(5):1–32.PubMed
3.
Zurück zum Zitat Thurman D, Guerrero J. Trends in hospitalization associated with traumatic brain injury. JAMA. 1999;282(10):954–7.PubMed Thurman D, Guerrero J. Trends in hospitalization associated with traumatic brain injury. JAMA. 1999;282(10):954–7.PubMed
5.
Zurück zum Zitat Smith DH, Uryu K, Saatman KE, Trojanowski JQ, McIntosh TK. Protein accumulation in traumatic brain injury. Neuromol Med. 2003;4(1–2):59–72. doi:10.1385/NMM:4:1-2:59. Smith DH, Uryu K, Saatman KE, Trojanowski JQ, McIntosh TK. Protein accumulation in traumatic brain injury. Neuromol Med. 2003;4(1–2):59–72. doi:10.​1385/​NMM:​4:​1-2:​59.
8.
Zurück zum Zitat Ponsford J, Willmott C, Rothwell A, Cameron P, Ayton G, Nelms R, et al. Cognitive and behavioral outcome following mild traumatic head injury in children. J Head Trauma Rehabil. 1999;14(4):360–72.PubMed Ponsford J, Willmott C, Rothwell A, Cameron P, Ayton G, Nelms R, et al. Cognitive and behavioral outcome following mild traumatic head injury in children. J Head Trauma Rehabil. 1999;14(4):360–72.PubMed
9.
Zurück zum Zitat Ponsford J, Willmott C, Rothwell A, Cameron P, Ayton G, Nelms R, et al. Impact of early intervention on outcome after mild traumatic brain injury in children. Pediatrics. 2001;108(6):1297–303.PubMed Ponsford J, Willmott C, Rothwell A, Cameron P, Ayton G, Nelms R, et al. Impact of early intervention on outcome after mild traumatic brain injury in children. Pediatrics. 2001;108(6):1297–303.PubMed
10.
Zurück zum Zitat Ponsford J, Cameron P, Fitzgerald M, Grant M, Mikocka-Walus A. Long-term outcomes after uncomplicated mild traumatic brain injury: a comparison with trauma controls. J Neurotrauma. 2011;28(6):937–46. doi:10.1089/neu.2010.1516.PubMed Ponsford J, Cameron P, Fitzgerald M, Grant M, Mikocka-Walus A. Long-term outcomes after uncomplicated mild traumatic brain injury: a comparison with trauma controls. J Neurotrauma. 2011;28(6):937–46. doi:10.​1089/​neu.​2010.​1516.PubMed
12.
Zurück zum Zitat Babikian T, Satz P, Zaucha K, Light R, Lewis RS, Asarnow RF. The UCLA longitudinal study of neurocognitive outcomes following mild pediatric traumatic brain injury. J Int Neuropsychol Soc. 2011;17:886–95. doi:10.1017/S1355617711000907. Babikian T, Satz P, Zaucha K, Light R, Lewis RS, Asarnow RF. The UCLA longitudinal study of neurocognitive outcomes following mild pediatric traumatic brain injury. J Int Neuropsychol Soc. 2011;17:886–95. doi:10.​1017/​S135561771100090​7.
13.
Zurück zum Zitat Kuppermann N, Holmes JF, Dayan PS, Hoyle Jr JD, Atabaki SM, Holubkov R, et al. Identification of children at very low risk of clinically-important brain injuries after head trauma: a prospective cohort study. Lancet. 2009;374(9696):1160–70. doi:10.1016/S0140-6736(09)61558-0.PubMed Kuppermann N, Holmes JF, Dayan PS, Hoyle Jr JD, Atabaki SM, Holubkov R, et al. Identification of children at very low risk of clinically-important brain injuries after head trauma: a prospective cohort study. Lancet. 2009;374(9696):1160–70. doi:10.​1016/​S0140-6736(09)61558-0.PubMed
14.
Zurück zum Zitat Schneier AJ, Shields BJ, Hostetler SG, Xiang H, Smith GA. Incidence of pediatric traumatic brain injury and associated hospital resource utilization in the United States. Pediatrics. 2006;118(2):483–92.PubMed Schneier AJ, Shields BJ, Hostetler SG, Xiang H, Smith GA. Incidence of pediatric traumatic brain injury and associated hospital resource utilization in the United States. Pediatrics. 2006;118(2):483–92.PubMed
15.
Zurück zum Zitat Brown AW, Leibson CL, Malec JF, Perkins PK, Diehl NN, Larson DR. Long-term survival after traumatic brain injury: a population-based analysis. NeuroRehabilitation. 2004;19(1):37–43.PubMed Brown AW, Leibson CL, Malec JF, Perkins PK, Diehl NN, Larson DR. Long-term survival after traumatic brain injury: a population-based analysis. NeuroRehabilitation. 2004;19(1):37–43.PubMed
16.
Zurück zum Zitat Harrison-Felix C, Whiteneck G, DeVivo M, Hammond FM, Jha A. Mortality following rehabilitation in the traumatic brain injury model systems of care. NeuroRehabilitation. 2004;19(1):45–54.PubMed Harrison-Felix C, Whiteneck G, DeVivo M, Hammond FM, Jha A. Mortality following rehabilitation in the traumatic brain injury model systems of care. NeuroRehabilitation. 2004;19(1):45–54.PubMed
17.
20.
23.
Zurück zum Zitat Satz P. Brain reserve capacity on symptom onset after brain injury: a formulation and review of evidence for threshold theory. Neuropsychology. 1993;7(3):273–95. Satz P. Brain reserve capacity on symptom onset after brain injury: a formulation and review of evidence for threshold theory. Neuropsychology. 1993;7(3):273–95.
24.
25.
Zurück zum Zitat DeKosky ST, Abrahamson EE, Ciallella JR, Paljug WR, Wisniewski SR, Clark RS, et al. Association of increased cortical soluble abeta42 levels with diffuse plaques after severe brain injury in humans. Arch Neurol. 2007;64(4):541–4. doi:10.1001/archneur.64.4.541.PubMed DeKosky ST, Abrahamson EE, Ciallella JR, Paljug WR, Wisniewski SR, Clark RS, et al. Association of increased cortical soluble abeta42 levels with diffuse plaques after severe brain injury in humans. Arch Neurol. 2007;64(4):541–4. doi:10.​1001/​archneur.​64.​4.​541.PubMed
27.
Zurück zum Zitat Ragan DK, McKinstry R, Benzinger T, Leonard J, Pineda JA. Depression of whole-brain oxygen extraction fraction is associated with poor outcome in pediatric traumatic brain injury. Pediatr Res. 2012;71(2):199–204. doi:10.1038/pr.2011.31.PubMedCentralPubMed Ragan DK, McKinstry R, Benzinger T, Leonard J, Pineda JA. Depression of whole-brain oxygen extraction fraction is associated with poor outcome in pediatric traumatic brain injury. Pediatr Res. 2012;71(2):199–204. doi:10.​1038/​pr.​2011.​31.PubMedCentralPubMed
28.
Zurück zum Zitat Cohen Z, Bonvento G, Lacombe P, Hamel E. Serotonin in the regulation of brain microcirculation. Prog Neurobiol. 1996;50(4):335–62.PubMed Cohen Z, Bonvento G, Lacombe P, Hamel E. Serotonin in the regulation of brain microcirculation. Prog Neurobiol. 1996;50(4):335–62.PubMed
31.
Zurück zum Zitat Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.PubMed Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41–53.PubMed
33.
Zurück zum Zitat Silverberg GD, Messier AA, Miller MC, Machan JT, Majmudar SS, Stopa EG, et al. Amyloid efflux transporter expression at the blood–brain barrier declines in normal aging. J Neuropathol Exp Neurol. 2010;69(10):1034–43. doi:10.1097/NEN.0b013e3181f46e25.PubMed Silverberg GD, Messier AA, Miller MC, Machan JT, Majmudar SS, Stopa EG, et al. Amyloid efflux transporter expression at the blood–brain barrier declines in normal aging. J Neuropathol Exp Neurol. 2010;69(10):1034–43. doi:10.​1097/​NEN.​0b013e3181f46e25​.PubMed
36.
Zurück zum Zitat Virgintino D, Robertson D, Errede M, Benagiano V, Tauer U, Roncali L, et al. Expression of caveolin-1 in human brain microvessels. Neuroscience. 2002;115(1):145–52.PubMed Virgintino D, Robertson D, Errede M, Benagiano V, Tauer U, Roncali L, et al. Expression of caveolin-1 in human brain microvessels. Neuroscience. 2002;115(1):145–52.PubMed
37.
Zurück zum Zitat Vogelgesang S, Warzok RW, Cascorbi I, Kunert-Keil C, Schroeder E, Kroemer HK, et al. The role of P-glycoprotein in cerebral amyloid angiopathy; implications for the early pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2004;1(2):121–5.PubMedCentralPubMed Vogelgesang S, Warzok RW, Cascorbi I, Kunert-Keil C, Schroeder E, Kroemer HK, et al. The role of P-glycoprotein in cerebral amyloid angiopathy; implications for the early pathogenesis of Alzheimer’s disease. Curr Alzheimer Res. 2004;1(2):121–5.PubMedCentralPubMed
38.
Zurück zum Zitat Ruderisch N, Virgintino D, Makrides V, Verrey F. Differential axial localization along the mouse brain vascular tree of luminal sodium-dependent glutamine transporters Snat1 and Snat3. J Cereb Blood Flow Metab. 2011;31(7):1637–47. doi:10.1038/jcbfm.2011.21.PubMedCentralPubMed Ruderisch N, Virgintino D, Makrides V, Verrey F. Differential axial localization along the mouse brain vascular tree of luminal sodium-dependent glutamine transporters Snat1 and Snat3. J Cereb Blood Flow Metab. 2011;31(7):1637–47. doi:10.​1038/​jcbfm.​2011.​21.PubMedCentralPubMed
39.
Zurück zum Zitat Saubamea B, Cochois-Guegan V, Cisternino S, Scherrmann JM. Heterogeneity in the rat brain vasculature revealed by quantitative confocal analysis of endothelial barrier antigen and P-glycoprotein expression. J Cereb Blood Flow Metab. 2012;32(1):81–92. doi:10.1038/jcbfm.2011.109.PubMedCentralPubMed Saubamea B, Cochois-Guegan V, Cisternino S, Scherrmann JM. Heterogeneity in the rat brain vasculature revealed by quantitative confocal analysis of endothelial barrier antigen and P-glycoprotein expression. J Cereb Blood Flow Metab. 2012;32(1):81–92. doi:10.​1038/​jcbfm.​2011.​109.PubMedCentralPubMed
40.
Zurück zum Zitat Badaut J, Nehlig A, Verbavatz J, Stoeckel M, Freund-Mercier MJ, Lasbennes F. Hypervascularization in the magnocellular nuclei of the rat hypothalamus: relationship with the distribution of aquaporin-4 and markers of energy metabolism. J Neuroendocrinol. 2000;12(10):960–9.PubMed Badaut J, Nehlig A, Verbavatz J, Stoeckel M, Freund-Mercier MJ, Lasbennes F. Hypervascularization in the magnocellular nuclei of the rat hypothalamus: relationship with the distribution of aquaporin-4 and markers of energy metabolism. J Neuroendocrinol. 2000;12(10):960–9.PubMed
44.
45.
Zurück zum Zitat Milner R, Campbell IL. Developmental regulation of beta1 integrins during angiogenesis in the central nervous system. Mol Cell Neurosci. 2002;20(4):616–26.PubMed Milner R, Campbell IL. Developmental regulation of beta1 integrins during angiogenesis in the central nervous system. Mol Cell Neurosci. 2002;20(4):616–26.PubMed
47.
Zurück zum Zitat Kim YS, Kim SS, Cho JJ, Choi DH, Hwang O, Shin DH, et al. Matrix metalloproteinase-3: a novel signaling proteinase from apoptotic neuronal cells that activates microglia. J Neurosci. 2005;25(14):3701–11. doi:10.1523/JNEUROSCI.4346-04.2005.PubMed Kim YS, Kim SS, Cho JJ, Choi DH, Hwang O, Shin DH, et al. Matrix metalloproteinase-3: a novel signaling proteinase from apoptotic neuronal cells that activates microglia. J Neurosci. 2005;25(14):3701–11. doi:10.​1523/​JNEUROSCI.​4346-04.​2005.PubMed
48.
Zurück zum Zitat Rafols JA, Kreipke CW, Petrov T. Alterations in cerebral cortex microvessels and the microcirculation in a rat model of traumatic brain injury: a correlative EM and laser Doppler flowmetry study. Neurol Res. 2007;29(4):339–47. doi:10.1179/016164107X204648.PubMed Rafols JA, Kreipke CW, Petrov T. Alterations in cerebral cortex microvessels and the microcirculation in a rat model of traumatic brain injury: a correlative EM and laser Doppler flowmetry study. Neurol Res. 2007;29(4):339–47. doi:10.​1179/​016164107X204648​.PubMed
49.
Zurück zum Zitat Badaut J, Moro V, Seylaz J, Lasbennes F. Distribution of muscarinic receptors on the endothelium of cortical vessels in the rat brain. Brain Res. 1997;778(1):25–33.PubMed Badaut J, Moro V, Seylaz J, Lasbennes F. Distribution of muscarinic receptors on the endothelium of cortical vessels in the rat brain. Brain Res. 1997;778(1):25–33.PubMed
53.
Zurück zum Zitat Li DY, Brooke B, Davis EC, Mecham RP, Sorensen LK, Boak BB, et al. Elastin is an essential determinant of arterial morphogenesis. Nature. 1998;393(6682):276–80. doi:10.1038/30522.PubMed Li DY, Brooke B, Davis EC, Mecham RP, Sorensen LK, Boak BB, et al. Elastin is an essential determinant of arterial morphogenesis. Nature. 1998;393(6682):276–80. doi:10.​1038/​30522.PubMed
54.
Zurück zum Zitat Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev. 1995;75(3):487–517.PubMed Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev. 1995;75(3):487–517.PubMed
55.
Zurück zum Zitat Rensen SS, Doevendans PA, van Eys GJ. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J Mon J Neth Soc Cardiol Neth Heart Found. 2007;15(3):100–8. Rensen SS, Doevendans PA, van Eys GJ. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J Mon J Neth Soc Cardiol Neth Heart Found. 2007;15(3):100–8.
56.
Zurück zum Zitat Hubbell MC, Semotiuk AJ, Thorpe RB, Adeoye OO, Butler SM, Williams JM, et al. Chronic hypoxia and VEGF differentially modulate abundance and organization of myosin heavy chain isoforms in fetal and adult ovine arteries. Am J Physiol. 2012;303:C1090–103. Hubbell MC, Semotiuk AJ, Thorpe RB, Adeoye OO, Butler SM, Williams JM, et al. Chronic hypoxia and VEGF differentially modulate abundance and organization of myosin heavy chain isoforms in fetal and adult ovine arteries. Am J Physiol. 2012;303:C1090–103.
57.
Zurück zum Zitat Baethmann A, Maier-Hauff K, Kempski O, Unterberg A, Wahl M, Schurer L. Mediators of brain edema and secondary brain damage. Crit Care Med. 1988;16(10):972–8.PubMed Baethmann A, Maier-Hauff K, Kempski O, Unterberg A, Wahl M, Schurer L. Mediators of brain edema and secondary brain damage. Crit Care Med. 1988;16(10):972–8.PubMed
58.
Zurück zum Zitat Sahuquillo J, Poca MA, Amoros S. Current aspects of pathophysiology and cell dysfunction after severe head injury. Curr Pharm Des. 2001;7(15):1475–503.PubMed Sahuquillo J, Poca MA, Amoros S. Current aspects of pathophysiology and cell dysfunction after severe head injury. Curr Pharm Des. 2001;7(15):1475–503.PubMed
59.
Zurück zum Zitat Gaetz M. The neurophysiology of brain injury. Clin Neurophysiol. 2004;115(1):4–18.PubMed Gaetz M. The neurophysiology of brain injury. Clin Neurophysiol. 2004;115(1):4–18.PubMed
60.
Zurück zum Zitat Zweckberger K, Eros C, Zimmermann R, Kim SW, Engel D, Plesnila N. Effect of early and delayed decompressive craniectomy on secondary brain damage after controlled cortical impact in mice. J Neurotrauma. 2006;23(7):1083–93. doi:10.1089/neu.2006.23.1083.PubMed Zweckberger K, Eros C, Zimmermann R, Kim SW, Engel D, Plesnila N. Effect of early and delayed decompressive craniectomy on secondary brain damage after controlled cortical impact in mice. J Neurotrauma. 2006;23(7):1083–93. doi:10.​1089/​neu.​2006.​23.​1083.PubMed
62.
63.
Zurück zum Zitat Bryan Jr RM, Cherian L, Robertson C. Regional cerebral blood flow after controlled cortical impact injury in rats. Anesth Analg. 1995;80(4):687–95.PubMed Bryan Jr RM, Cherian L, Robertson C. Regional cerebral blood flow after controlled cortical impact injury in rats. Anesth Analg. 1995;80(4):687–95.PubMed
64.
Zurück zum Zitat Engel DC, Mies G, Terpolilli NA, Trabold R, Loch A, De Zeeuw CI, et al. Changes of cerebral blood flow during the secondary expansion of a cortical contusion assessed by 14C-iodoantipyrine autoradiography in mice using a non-invasive protocol. J Neurotrauma. 2008;25(7):739–53. doi:10.1089/neu.2007.0480.PubMed Engel DC, Mies G, Terpolilli NA, Trabold R, Loch A, De Zeeuw CI, et al. Changes of cerebral blood flow during the secondary expansion of a cortical contusion assessed by 14C-iodoantipyrine autoradiography in mice using a non-invasive protocol. J Neurotrauma. 2008;25(7):739–53. doi:10.​1089/​neu.​2007.​0480.PubMed
65.
Zurück zum Zitat Kochanek PM, Marion DW, Zhang W, Schiding JK, White M, Palmer AM, et al. Severe controlled cortical impact in rats: assessment of cerebral edema, blood flow, and contusion volume. J Neurotrauma. 1995;12(6):1015–25.PubMed Kochanek PM, Marion DW, Zhang W, Schiding JK, White M, Palmer AM, et al. Severe controlled cortical impact in rats: assessment of cerebral edema, blood flow, and contusion volume. J Neurotrauma. 1995;12(6):1015–25.PubMed
66.
Zurück zum Zitat Schroder ML, Muizelaar JP, Bullock MR, Salvant JB, Povlishock JT. Focal ischemia due to traumatic contusions documented by stable xenon-CT and ultrastructural studies. J Neurosurg. 1995;82(6):966–71. doi:10.3171/jns.1995.82.6.0966.PubMed Schroder ML, Muizelaar JP, Bullock MR, Salvant JB, Povlishock JT. Focal ischemia due to traumatic contusions documented by stable xenon-CT and ultrastructural studies. J Neurosurg. 1995;82(6):966–71. doi:10.​3171/​jns.​1995.​82.​6.​0966.PubMed
68.
Zurück zum Zitat Sahuquillo J, Munar F, Baguena M, Poca MA, Pedraza S, Rodriguez-Baeza A. Evaluation of cerebrovascular CO2-reactivity and autoregulation in patients with post-traumatic diffuse brain swelling (diffuse injury III). Acta Neurochir Suppl. 1998;71:233–6.PubMed Sahuquillo J, Munar F, Baguena M, Poca MA, Pedraza S, Rodriguez-Baeza A. Evaluation of cerebrovascular CO2-reactivity and autoregulation in patients with post-traumatic diffuse brain swelling (diffuse injury III). Acta Neurochir Suppl. 1998;71:233–6.PubMed
69.
Zurück zum Zitat Vavilala MS, Muangman S, Tontisirin N, Fisk D, Roscigno C, Mitchell P, et al. Impaired cerebral autoregulation and 6-month outcome in children with severe traumatic brain injury: preliminary findings. Dev Neurosci. 2006;28(4–5):348–53. doi:10.1159/000094161.PubMed Vavilala MS, Muangman S, Tontisirin N, Fisk D, Roscigno C, Mitchell P, et al. Impaired cerebral autoregulation and 6-month outcome in children with severe traumatic brain injury: preliminary findings. Dev Neurosci. 2006;28(4–5):348–53. doi:10.​1159/​000094161.PubMed
70.
Zurück zum Zitat Muizelaar JP. The use of electroencephalography and brain protection during operation for basilar aneurysms. Neurosurgery. 1989;25(6):899–903.PubMed Muizelaar JP. The use of electroencephalography and brain protection during operation for basilar aneurysms. Neurosurgery. 1989;25(6):899–903.PubMed
71.
Zurück zum Zitat Muizelaar JP, Ward JD, Marmarou A, Newlon PG, Wachi A. Cerebral blood flow and metabolism in severely head-injured children. Part 2: autoregulation. J Neurosurg. 1989;71(1):72–6.PubMed Muizelaar JP, Ward JD, Marmarou A, Newlon PG, Wachi A. Cerebral blood flow and metabolism in severely head-injured children. Part 2: autoregulation. J Neurosurg. 1989;71(1):72–6.PubMed
72.
Zurück zum Zitat Freeman SS, Udomphorn Y, Armstead WM, Fisk DM, Vavilala MS. Young age as a risk factor for impaired cerebral autoregulation after moderate to severe pediatric traumatic brain injury. Anesthesiology. 2008;108(4):588–95. doi:10.1097/ALN.0b013e31816725d7.PubMed Freeman SS, Udomphorn Y, Armstead WM, Fisk DM, Vavilala MS. Young age as a risk factor for impaired cerebral autoregulation after moderate to severe pediatric traumatic brain injury. Anesthesiology. 2008;108(4):588–95. doi:10.​1097/​ALN.​0b013e31816725d7​.PubMed
73.
Zurück zum Zitat Sharples PM, Stuart AG, Matthews DS, Aynsley-Green A, Eyre JA. Cerebral blood flow and metabolism in children with severe head injury. Part 1: relation to age, Glasgow coma score, outcome, intracranial pressure, and time after injury. J Neurol Neurosurg Psychiatry. 1995;58(2):145–52.PubMedCentralPubMed Sharples PM, Stuart AG, Matthews DS, Aynsley-Green A, Eyre JA. Cerebral blood flow and metabolism in children with severe head injury. Part 1: relation to age, Glasgow coma score, outcome, intracranial pressure, and time after injury. J Neurol Neurosurg Psychiatry. 1995;58(2):145–52.PubMedCentralPubMed
74.
Zurück zum Zitat Sharples PM, Matthews DS, Eyre JA. Cerebral blood flow and metabolism in children with severe head injuries. Part 2: cerebrovascular resistance and its determinants. J Neurol Neurosurg Psychiatry. 1995;58(2):153–9.PubMedCentralPubMed Sharples PM, Matthews DS, Eyre JA. Cerebral blood flow and metabolism in children with severe head injuries. Part 2: cerebrovascular resistance and its determinants. J Neurol Neurosurg Psychiatry. 1995;58(2):153–9.PubMedCentralPubMed
75.
Zurück zum Zitat Armstead WM. Cerebral hemodynamics after traumatic brain injury of immature brain. Exp Toxicol Pathol. 1999;51(2):137–42.PubMed Armstead WM. Cerebral hemodynamics after traumatic brain injury of immature brain. Exp Toxicol Pathol. 1999;51(2):137–42.PubMed
76.
Zurück zum Zitat Ashwal S, Holshouser BA, Shu SK, Simmons PL, Perkin RM, Tomasi LG, et al. Predictive value of proton magnetic resonance spectroscopy in pediatric closed head injury. Pediatr Neurol. 2000;23(2):114–25.PubMed Ashwal S, Holshouser BA, Shu SK, Simmons PL, Perkin RM, Tomasi LG, et al. Predictive value of proton magnetic resonance spectroscopy in pediatric closed head injury. Pediatr Neurol. 2000;23(2):114–25.PubMed
77.
Zurück zum Zitat Bartnik BL, Sutton RL, Fukushima M, Harris NG, Hovda DA, Lee SM. Upregulation of pentose phosphate pathway and preservation of tricarboxylic acid cycle flux after experimental brain injury. J Neurotrauma. 2005;22(10):1052–65. doi:10.1089/neu.2005.22.1052.PubMed Bartnik BL, Sutton RL, Fukushima M, Harris NG, Hovda DA, Lee SM. Upregulation of pentose phosphate pathway and preservation of tricarboxylic acid cycle flux after experimental brain injury. J Neurotrauma. 2005;22(10):1052–65. doi:10.​1089/​neu.​2005.​22.​1052.PubMed
79.
Zurück zum Zitat Ashwal S, Holshouser B, Tong K, Serna T, Osterdock R, Gross M, et al. Proton MR spectroscopy detected glutamate/glutamine is increased in children with traumatic brain injury. J Neurotrauma. 2004;21(11):1539–52.PubMed Ashwal S, Holshouser B, Tong K, Serna T, Osterdock R, Gross M, et al. Proton MR spectroscopy detected glutamate/glutamine is increased in children with traumatic brain injury. J Neurotrauma. 2004;21(11):1539–52.PubMed
80.
Zurück zum Zitat Armstead WM. Age-dependent impairment of K(ATP) channel function following brain injury. J Neurotrauma. 1999;16(5):391–402.PubMed Armstead WM. Age-dependent impairment of K(ATP) channel function following brain injury. J Neurotrauma. 1999;16(5):391–402.PubMed
82.
Zurück zum Zitat Cherian L, Hlatky R, Robertson CS. Nitric oxide in traumatic brain injury. Brain Pathol. 2004;14(2):195–201.PubMed Cherian L, Hlatky R, Robertson CS. Nitric oxide in traumatic brain injury. Brain Pathol. 2004;14(2):195–201.PubMed
85.
Zurück zum Zitat Hlatky R, Lui H, Cherian L, Goodman JC, O’Brien WE, Contant CF, et al. The role of endothelial nitric oxide synthase in the cerebral hemodynamics after controlled cortical impact injury in mice. J Neurotrauma. 2003;20(10):995–1006. doi:10.1089/089771503770195849.PubMed Hlatky R, Lui H, Cherian L, Goodman JC, O’Brien WE, Contant CF, et al. The role of endothelial nitric oxide synthase in the cerebral hemodynamics after controlled cortical impact injury in mice. J Neurotrauma. 2003;20(10):995–1006. doi:10.​1089/​0897715037701958​49.PubMed
86.
Zurück zum Zitat Cherian L, Chacko G, Goodman C, Robertson CS. Neuroprotective effects of L-arginine administration after cortical impact injury in rats: dose response and time window. J Pharmacol Exp Ther. 2003;304(2):617–23. doi:10.1124/jpet.102.043430.PubMed Cherian L, Chacko G, Goodman C, Robertson CS. Neuroprotective effects of L-arginine administration after cortical impact injury in rats: dose response and time window. J Pharmacol Exp Ther. 2003;304(2):617–23. doi:10.​1124/​jpet.​102.​043430.PubMed
87.
Zurück zum Zitat Orihara Y, Ikematsu K, Tsuda R, Nakasono I. Induction of nitric oxide synthase by traumatic brain injury. Forensic Sci Int. 2001;123(2–3):142–9.PubMed Orihara Y, Ikematsu K, Tsuda R, Nakasono I. Induction of nitric oxide synthase by traumatic brain injury. Forensic Sci Int. 2001;123(2–3):142–9.PubMed
88.
Zurück zum Zitat Steiner J, Rafols D, Park HK, Katar MS, Rafols JA, Petrov T. Attenuation of iNOS mRNA exacerbates hypoperfusion and upregulates endothelin-1 expression in hippocampus and cortex after brain trauma. Nitric Oxide. 2004;10(3):162–9. doi:10.1016/j.niox.2004.03.005.PubMed Steiner J, Rafols D, Park HK, Katar MS, Rafols JA, Petrov T. Attenuation of iNOS mRNA exacerbates hypoperfusion and upregulates endothelin-1 expression in hippocampus and cortex after brain trauma. Nitric Oxide. 2004;10(3):162–9. doi:10.​1016/​j.​niox.​2004.​03.​005.PubMed
89.
Zurück zum Zitat Xia Y, Zweier JL. Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc Natl Acad Sci U S A. 1997;94(13):6954–8.PubMedCentralPubMed Xia Y, Zweier JL. Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc Natl Acad Sci U S A. 1997;94(13):6954–8.PubMedCentralPubMed
90.
Zurück zum Zitat Xia Y, Tsai AL, Berka V, Zweier JL. Superoxide generation from endothelial nitric-oxide synthase. A Ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J Biol Chem. 1998;273(40):25804–8.PubMed Xia Y, Tsai AL, Berka V, Zweier JL. Superoxide generation from endothelial nitric-oxide synthase. A Ca2+/calmodulin-dependent and tetrahydrobiopterin regulatory process. J Biol Chem. 1998;273(40):25804–8.PubMed
92.
Zurück zum Zitat Gu Y, Zheng G, Xu M, Li Y, Chen X, Zhu W, et al. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood–brain barrier permeability in focal cerebral ischemia and reperfusion injury. J Neurochem. 2011. doi:10.1111/j.1471-4159.2011.07542.x. Gu Y, Zheng G, Xu M, Li Y, Chen X, Zhu W, et al. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood–brain barrier permeability in focal cerebral ischemia and reperfusion injury. J Neurochem. 2011. doi:10.​1111/​j.​1471-4159.​2011.​07542.​x.
95.
Zurück zum Zitat Plesnila N, Friedrich D, Eriskat J, Baethmann A, Stoffel M. Relative cerebral blood flow during the secondary expansion of a cortical lesion in rats. Neurosci Lett. 2003;345(2):85–8.PubMed Plesnila N, Friedrich D, Eriskat J, Baethmann A, Stoffel M. Relative cerebral blood flow during the secondary expansion of a cortical lesion in rats. Neurosci Lett. 2003;345(2):85–8.PubMed
96.
Zurück zum Zitat Armstead WM. Brain injury impairs ATP-sensitive K+ channel function in piglet cerebral arteries. Stroke. 1997;28(11):2273–9. discussion 80.PubMed Armstead WM. Brain injury impairs ATP-sensitive K+ channel function in piglet cerebral arteries. Stroke. 1997;28(11):2273–9. discussion 80.PubMed
97.
Zurück zum Zitat Kontos HA, Wei EP. Endothelium-dependent responses after experimental brain injury. J Neurotrauma. 1992;9(4):349–54.PubMed Kontos HA, Wei EP. Endothelium-dependent responses after experimental brain injury. J Neurotrauma. 1992;9(4):349–54.PubMed
98.
99.
Zurück zum Zitat Sercombe R, Dinh YR, Gomis P. Cerebrovascular inflammation following subarachnoid hemorrhage. Jpn J Pharmacol. 2002;88(3):227–49.PubMed Sercombe R, Dinh YR, Gomis P. Cerebrovascular inflammation following subarachnoid hemorrhage. Jpn J Pharmacol. 2002;88(3):227–49.PubMed
100.
Zurück zum Zitat Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, et al. Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci. 2000;20(18):7037–42.PubMed Wang X, Jung J, Asahi M, Chwang W, Russo L, Moskowitz MA, et al. Effects of matrix metalloproteinase-9 gene knock-out on morphological and motor outcomes after traumatic brain injury. J Neurosci. 2000;20(18):7037–42.PubMed
102.
103.
Zurück zum Zitat Roberts DJ, Jenne CN, Leger C, Kramer AH, Gallagher CN, Todd S, et al. A prospective evaluation of the temporal matrix metalloproteinase response after severe traumatic brain injury in humans. J Neurotrauma. 2013. doi:10.1089/neu.2012.2841. Roberts DJ, Jenne CN, Leger C, Kramer AH, Gallagher CN, Todd S, et al. A prospective evaluation of the temporal matrix metalloproteinase response after severe traumatic brain injury in humans. J Neurotrauma. 2013. doi:10.​1089/​neu.​2012.​2841.
104.
Zurück zum Zitat Suehiro E, Fujisawa H, Akimura T, Ishihara H, Kajiwara K, Kato S, et al. Increased matrix metalloproteinase-9 in blood in association with activation of interleukin-6 after traumatic brain injury: influence of hypothermic therapy. J Neurotrauma. 2004;21(12):1706–11. doi:10.1089/neu.2004.21.1706.PubMed Suehiro E, Fujisawa H, Akimura T, Ishihara H, Kajiwara K, Kato S, et al. Increased matrix metalloproteinase-9 in blood in association with activation of interleukin-6 after traumatic brain injury: influence of hypothermic therapy. J Neurotrauma. 2004;21(12):1706–11. doi:10.​1089/​neu.​2004.​21.​1706.PubMed
105.
Zurück zum Zitat Beaumont A, Fatouros P, Gennarelli T, Corwin F, Marmarou A. Bolus tracer delivery measured by MRI confirms edema without blood–brain barrier permeability in diffuse traumatic brain injury. Acta Neurochir Suppl. 2006;96:171–4.PubMed Beaumont A, Fatouros P, Gennarelli T, Corwin F, Marmarou A. Bolus tracer delivery measured by MRI confirms edema without blood–brain barrier permeability in diffuse traumatic brain injury. Acta Neurochir Suppl. 2006;96:171–4.PubMed
106.
Zurück zum Zitat Nag S, Venugopalan R, Stewart DJ. Increased caveolin-1 expression precedes decreased expression of occludin and claudin-5 during blood–brain barrier breakdown. Acta Neuropathol. 2007;114(5):459–69. doi:10.1007/s00401-007-0274-x.PubMed Nag S, Venugopalan R, Stewart DJ. Increased caveolin-1 expression precedes decreased expression of occludin and claudin-5 during blood–brain barrier breakdown. Acta Neuropathol. 2007;114(5):459–69. doi:10.​1007/​s00401-007-0274-x.PubMed
107.
Zurück zum Zitat Pierre K, Pellerin L. Monocarboxylate transporters in the central nervous system: distribution, regulation and function. J Neurochem. 2005;94(1):1–14.PubMed Pierre K, Pellerin L. Monocarboxylate transporters in the central nervous system: distribution, regulation and function. J Neurochem. 2005;94(1):1–14.PubMed
108.
Zurück zum Zitat Prins ML, Giza CC. Induction of monocarboxylate transporter 2 expression and ketone transport following traumatic brain injury in juvenile and adult rats. Dev Neurosci. 2006;28(4–5):447–56.PubMed Prins ML, Giza CC. Induction of monocarboxylate transporter 2 expression and ketone transport following traumatic brain injury in juvenile and adult rats. Dev Neurosci. 2006;28(4–5):447–56.PubMed
110.
111.
Zurück zum Zitat Abrahamson EE, Foley LM, Dekosky ST, Kevin Hitchens T, Ho C, Kochanek PM, et al. Cerebral blood flow changes after brain injury in human amyloid-beta knock-in mice. J Cereb Blood Flow Metab. 2013;33(6):826–33. doi:10.1038/jcbfm.2013.24.PubMed Abrahamson EE, Foley LM, Dekosky ST, Kevin Hitchens T, Ho C, Kochanek PM, et al. Cerebral blood flow changes after brain injury in human amyloid-beta knock-in mice. J Cereb Blood Flow Metab. 2013;33(6):826–33. doi:10.​1038/​jcbfm.​2013.​24.PubMed
113.
Zurück zum Zitat Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, et al. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci. 2011;12(3):169–82. doi:10.1038/nrn2995.PubMedCentralPubMed Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, et al. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci. 2011;12(3):169–82. doi:10.​1038/​nrn2995.PubMedCentralPubMed
114.
Zurück zum Zitat Strbian D, Durukan A, Pitkonen M, Marinkovic I, Tatlisumak E, Pedrono E, et al. The blood–brain barrier is continuously open for several weeks following transient focal cerebral ischemia. Neuroscience. 2008;153(1):175–81.PubMed Strbian D, Durukan A, Pitkonen M, Marinkovic I, Tatlisumak E, Pedrono E, et al. The blood–brain barrier is continuously open for several weeks following transient focal cerebral ischemia. Neuroscience. 2008;153(1):175–81.PubMed
115.
Zurück zum Zitat Pop V, Sorensen DW, Kamper JE, Ajao DO, Murphy MP, Head E, et al. Early brain injury alters the blood–brain barrier phenotype in parallel with beta-amyloid and cognitive changes in adulthood. J Cereb Blood Flow Metab. 2013;33(2):205–14. doi:10.1038/jcbfm.2012.154.PubMedCentralPubMed Pop V, Sorensen DW, Kamper JE, Ajao DO, Murphy MP, Head E, et al. Early brain injury alters the blood–brain barrier phenotype in parallel with beta-amyloid and cognitive changes in adulthood. J Cereb Blood Flow Metab. 2013;33(2):205–14. doi:10.​1038/​jcbfm.​2012.​154.PubMedCentralPubMed
116.
Zurück zum Zitat Lin JL, Huang YH, Shen YC, Huang HC, Liu PH. Ascorbic acid prevents blood–brain barrier disruption and sensory deficit caused by sustained compression of primary somatosensory cortex. J Cereb Blood Flow Metab. 2010;30(6):1121–36. doi:10.1038/jcbfm.2009.277.PubMedCentralPubMed Lin JL, Huang YH, Shen YC, Huang HC, Liu PH. Ascorbic acid prevents blood–brain barrier disruption and sensory deficit caused by sustained compression of primary somatosensory cortex. J Cereb Blood Flow Metab. 2010;30(6):1121–36. doi:10.​1038/​jcbfm.​2009.​277.PubMedCentralPubMed
117.
Zurück zum Zitat Cirrito JR, Deane R, Fagan AM, Spinner ML, Parsadanian M, Finn MB, et al. P-glycoprotein deficiency at the blood–brain barrier increases amyloid-beta deposition in an Alzheimer disease mouse model. J Clin Invest. 2005;115(11):3285–90. doi:10.1172/JCI25247.PubMedCentralPubMed Cirrito JR, Deane R, Fagan AM, Spinner ML, Parsadanian M, Finn MB, et al. P-glycoprotein deficiency at the blood–brain barrier increases amyloid-beta deposition in an Alzheimer disease mouse model. J Clin Invest. 2005;115(11):3285–90. doi:10.​1172/​JCI25247.PubMedCentralPubMed
120.
Zurück zum Zitat Jodoin J, Demeule M, Fenart L, Cecchelli R, Farmer S, Linton KJ, et al. P-glycoprotein in blood–brain barrier endothelial cells: interaction and oligomerization with caveolins. J Neurochem. 2003;87(4):1010–23.PubMed Jodoin J, Demeule M, Fenart L, Cecchelli R, Farmer S, Linton KJ, et al. P-glycoprotein in blood–brain barrier endothelial cells: interaction and oligomerization with caveolins. J Neurochem. 2003;87(4):1010–23.PubMed
121.
Zurück zum Zitat Predescu D, Palade GE. Plasmalemmal vesicles represent the large pore system of continuous microvascular endothelium. Am J Physiol. 1993;265(2 Pt 2):H725–33.PubMed Predescu D, Palade GE. Plasmalemmal vesicles represent the large pore system of continuous microvascular endothelium. Am J Physiol. 1993;265(2 Pt 2):H725–33.PubMed
122.
Zurück zum Zitat Lisanti MP, Scherer PE, Tang Z, Sargiacomo M. Caveolae, caveolin and caveolin-rich membrane domains: a signalling hypothesis. Trends Cell Biol. 1994;4(7):231–5.PubMed Lisanti MP, Scherer PE, Tang Z, Sargiacomo M. Caveolae, caveolin and caveolin-rich membrane domains: a signalling hypothesis. Trends Cell Biol. 1994;4(7):231–5.PubMed
123.
Zurück zum Zitat Bucci M, Gratton JP, Rudic RD, Acevedo L, Roviezzo F, Cirino G, et al. In vivo delivery of the caveolin-1 scaffolding domain inhibits nitric oxide synthesis and reduces inflammation. Nat Med. 2000;6(12):1362–7. doi:10.1038/82176.PubMed Bucci M, Gratton JP, Rudic RD, Acevedo L, Roviezzo F, Cirino G, et al. In vivo delivery of the caveolin-1 scaffolding domain inhibits nitric oxide synthesis and reduces inflammation. Nat Med. 2000;6(12):1362–7. doi:10.​1038/​82176.PubMed
124.
Zurück zum Zitat Bauer PM, Yu J, Chen Y, Hickey R, Bernatchez PN, Looft-Wilson R, et al. Endothelial-specific expression of caveolin-1 impairs microvascular permeability and angiogenesis. Proc Natl Acad Sci U S A. 2005;102(1):204–9. doi:10.1073/pnas.0406092102.PubMedCentralPubMed Bauer PM, Yu J, Chen Y, Hickey R, Bernatchez PN, Looft-Wilson R, et al. Endothelial-specific expression of caveolin-1 impairs microvascular permeability and angiogenesis. Proc Natl Acad Sci U S A. 2005;102(1):204–9. doi:10.​1073/​pnas.​0406092102.PubMedCentralPubMed
127.
Zurück zum Zitat McCaffrey G, Staatz WD, Quigley CA, Nametz N, Seelbach MJ, Campos CR, et al. Tight junctions contain oligomeric protein assembly critical for maintaining blood–brain barrier integrity in vivo. J Neurochem. 2007;103(6):2540–55. doi:10.1111/j.1471-4159.2007.04943.x.PubMed McCaffrey G, Staatz WD, Quigley CA, Nametz N, Seelbach MJ, Campos CR, et al. Tight junctions contain oligomeric protein assembly critical for maintaining blood–brain barrier integrity in vivo. J Neurochem. 2007;103(6):2540–55. doi:10.​1111/​j.​1471-4159.​2007.​04943.​x.PubMed
128.
Zurück zum Zitat McCaffrey G, Staatz WD, Sanchez-Covarrubias L, Finch JD, Demarco K, Laracuente ML, et al. P-glycoprotein trafficking at the blood–brain barrier altered by peripheral inflammatory hyperalgesia. J Neurochem. 2012;122(5):962–75. doi:10.1111/j.1471-4159.2012.07831.x.PubMed McCaffrey G, Staatz WD, Sanchez-Covarrubias L, Finch JD, Demarco K, Laracuente ML, et al. P-glycoprotein trafficking at the blood–brain barrier altered by peripheral inflammatory hyperalgesia. J Neurochem. 2012;122(5):962–75. doi:10.​1111/​j.​1471-4159.​2012.​07831.​x.PubMed
129.
Zurück zum Zitat Mellergard P, Sjogren F, Hillman J. Release of VEGF and FGF in the extracellular space following severe subarachnoidal haemorrhage or traumatic head injury in humans. Br J Neurosurg. 2010;24(3):261–7. doi:10.3109/02688690903521605.PubMed Mellergard P, Sjogren F, Hillman J. Release of VEGF and FGF in the extracellular space following severe subarachnoidal haemorrhage or traumatic head injury in humans. Br J Neurosurg. 2010;24(3):261–7. doi:10.​3109/​0268869090352160​5.PubMed
130.
Zurück zum Zitat Morgan R, Kreipke CW, Roberts G, Bagchi M, Rafols JA. Neovascularization following traumatic brain injury: possible evidence for both angiogenesis and vasculogenesis. Neurol Res. 2007;29(4):375–81. doi:10.1179/016164107X204693.PubMed Morgan R, Kreipke CW, Roberts G, Bagchi M, Rafols JA. Neovascularization following traumatic brain injury: possible evidence for both angiogenesis and vasculogenesis. Neurol Res. 2007;29(4):375–81. doi:10.​1179/​016164107X204693​.PubMed
Metadaten
Titel
Vascular Neural Network Phenotypic Transformation After Traumatic Injury: Potential Role in Long-Term Sequelae
verfasst von
J. Badaut
G. J. Bix
Publikationsdatum
01.06.2014
Verlag
Springer US
Erschienen in
Translational Stroke Research / Ausgabe 3/2014
Print ISSN: 1868-4483
Elektronische ISSN: 1868-601X
DOI
https://doi.org/10.1007/s12975-013-0304-z

Weitere Artikel der Ausgabe 3/2014

Translational Stroke Research 3/2014 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.