Skip to main content
Top
Published in: Molecular Neurodegeneration 1/2018

Open Access 01-12-2018 | Research article

Apolipoprotein E4 impairs spontaneous blood brain barrier repair following traumatic brain injury

Authors: Bevan S. Main, Sonia Villapol, Stephanie S. Sloley, David J. Barton, Maia Parsadanian, Chinyere Agbaegbu, Kathryn Stefos, Mondona S. McCann, Patricia M. Washington, Olga C. Rodriguez, Mark P. Burns

Published in: Molecular Neurodegeneration | Issue 1/2018

Login to get access

Abstract

Background

Traumatic Brain Injury (TBI) is a major cause of disability and mortality, to which there is currently no comprehensive treatment. Blood Brain Barrier (BBB) dysfunction is well documented in human TBI patients, yet the molecular mechanisms that underlie this neurovascular unit (NVU) pathology remains unclear. The apolipoprotein-E (apoE) protein has been implicated in controlling BBB integrity in an isoform dependent manner, via suppression of Cyclophilin A (CypA)–Matrix metallopeptidase-9 (MMP-9) signaling cascades, however the contribution of this pathway in TBI-induced BBB permeability is not fully investigated.

Methods

We exposed C57Bl/6 mice to controlled cortical impact and assessed NVU and BBB permeability responses up to 21 days post-injury. We pharmacologically probed the role of the CypA-MMP-9 pathway in BBB permeability after TBI using Cyclosporin A (CsA, 20 mg/kg). Finally, as the apoE4 protein is known to be functionally deficient compared to the apoE3 protein, we used humanized APOE mice as a clinically relevant model to study the role of apoE on BBB injury and repair after TBI.

Results

In C57Bl/6 mice there was an inverse relationship between soluble apoE and BBB permeability, such that damaged BBB stabilizes as apoE levels increase in the days following TBI. TBI mice displayed acute pericyte loss, increased MMP-9 production and activity, and reduced tight-junction expression. Treatment with the CypA antagonist CsA in C57Bl/6 mice attenuates MMP-9 responses and enhances BBB repair after injury, demonstrating that MMP-9 plays an important role in the timing of spontaneous BBB repair after TBI. We also show that apoe mRNA is present in both astrocytes and pericytes after TBI.
We report that APOE3 and APOE4 mice have similar acute BBB responses to TBI, but APOE3 mice display faster spontaneous BBB repair than APOE4 mice. Isolated microvessel analysis reveals delayed pericyte repopulation, augmented and sustained MMP-9 expression at the NVU, and impaired stabilization of Zonula Occludens-1, Occludin and Claudin-5 expression at tight junctions in APOE4 mice after TBI compared to APOE3 mice.

Conclusions

These data confirm apoE as an important modulator of spontaneous BBB stabilization following TBI, and highlights the APOE4 allele as a risk factor for poor outcome after TBI.
Literature
1.
go back to reference Neuwelt E, et al. Strategies to advance translational research into brain barriers. The Lancet Neurology. 2008;7(1):84–96.CrossRefPubMed Neuwelt E, et al. Strategies to advance translational research into brain barriers. The Lancet Neurology. 2008;7(1):84–96.CrossRefPubMed
3.
go back to reference Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240(4852):622–30.CrossRefPubMed Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science. 1988;240(4852):622–30.CrossRefPubMed
4.
go back to reference Chen Y, et al. Motor and cognitive deficits in apolipoprotein E-deficient mice after closed head injury. Neuroscience. 1997;80(4):1255–62.CrossRefPubMed Chen Y, et al. Motor and cognitive deficits in apolipoprotein E-deficient mice after closed head injury. Neuroscience. 1997;80(4):1255–62.CrossRefPubMed
5.
go back to reference Namjoshi DR, et al. The liver X receptor agonist GW3965 improves recovery from mild repetitive traumatic brain injury in mice partly through apolipoprotein E. PLoS One. 2013;8(1):e53529.CrossRefPubMedPubMedCentral Namjoshi DR, et al. The liver X receptor agonist GW3965 improves recovery from mild repetitive traumatic brain injury in mice partly through apolipoprotein E. PLoS One. 2013;8(1):e53529.CrossRefPubMedPubMedCentral
6.
go back to reference Horsburgh K, et al. Intraventricular infusion of apolipoprotein E ameliorates acute neuronal damage after global cerebral ischemia in mice. J Cereb Blood Flow Metab. 2000;20(3):458–62.CrossRefPubMed Horsburgh K, et al. Intraventricular infusion of apolipoprotein E ameliorates acute neuronal damage after global cerebral ischemia in mice. J Cereb Blood Flow Metab. 2000;20(3):458–62.CrossRefPubMed
7.
go back to reference Kay AD, et al. Alterations in cerebrospinal fluid apolipoprotein E and amyloid beta-protein after traumatic brain injury. J Neurotrauma. 2003;20(10):943–52.CrossRefPubMed Kay AD, et al. Alterations in cerebrospinal fluid apolipoprotein E and amyloid beta-protein after traumatic brain injury. J Neurotrauma. 2003;20(10):943–52.CrossRefPubMed
8.
go back to reference Washington PM, Burns MP. The effect of the APOE4 gene on accumulation of Abeta40 after brain injury cannot be reversed by increasing apoE4 protein. J Neuropathol Exp Neurol. 2016; Washington PM, Burns MP. The effect of the APOE4 gene on accumulation of Abeta40 after brain injury cannot be reversed by increasing apoE4 protein. J Neuropathol Exp Neurol. 2016;
10.
go back to reference Methia N, et al. ApoE deficiency compromises the blood brain barrier especially after injury. Mol Med. 2001;7(12):810–5.PubMedPubMedCentral Methia N, et al. ApoE deficiency compromises the blood brain barrier especially after injury. Mol Med. 2001;7(12):810–5.PubMedPubMedCentral
11.
go back to reference Hafezi-Moghadam A, Thomas KL, Wagner DD. ApoE deficiency leads to a progressive age-dependent blood-brain barrier leakage. Am J Physiol Cell Physiol. 2007;292(4):C1256–62.CrossRefPubMed Hafezi-Moghadam A, Thomas KL, Wagner DD. ApoE deficiency leads to a progressive age-dependent blood-brain barrier leakage. Am J Physiol Cell Physiol. 2007;292(4):C1256–62.CrossRefPubMed
12.
go back to reference Jordan B. Apolipoprotein E ∈4 associated with chronic traumatic brain injury in boxing. JAMA. 1997;278(2):136.CrossRefPubMed Jordan B. Apolipoprotein E ∈4 associated with chronic traumatic brain injury in boxing. JAMA. 1997;278(2):136.CrossRefPubMed
13.
go back to reference Teasdale G, et al. Association of apolipoprotein E polymorphism with outcome after head injury. Lancet. 1997;350(9084):1069–71.CrossRefPubMed Teasdale G, et al. Association of apolipoprotein E polymorphism with outcome after head injury. Lancet. 1997;350(9084):1069–71.CrossRefPubMed
14.
15.
go back to reference Friedman G, et al. Apolipoprotein E-epsilon4 genotype predicts a poor outcome in survivors of traumatic brain injury. Neurology. 1999;52(2):244–8.CrossRefPubMed Friedman G, et al. Apolipoprotein E-epsilon4 genotype predicts a poor outcome in survivors of traumatic brain injury. Neurology. 1999;52(2):244–8.CrossRefPubMed
16.
go back to reference Mayeux R, et al. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer's disease. Neurology. 1995;45(3 Pt 1):555–7.CrossRefPubMed Mayeux R, et al. Synergistic effects of traumatic head injury and apolipoprotein-epsilon 4 in patients with Alzheimer's disease. Neurology. 1995;45(3 Pt 1):555–7.CrossRefPubMed
17.
go back to reference Sabo T, et al. Susceptibility of transgenic mice expressing human apolipoprotein E to closed head injury: the allele E3 is neuroprotective whereas E4 increases fatalities. Neuroscience. 2000;101(4):879–84.CrossRefPubMed Sabo T, et al. Susceptibility of transgenic mice expressing human apolipoprotein E to closed head injury: the allele E3 is neuroprotective whereas E4 increases fatalities. Neuroscience. 2000;101(4):879–84.CrossRefPubMed
18.
go back to reference Mannix R, et al. Age-dependent effect of apolipoprotein E4 on functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab. 2011;31(1):351–61.CrossRefPubMed Mannix R, et al. Age-dependent effect of apolipoprotein E4 on functional outcome after controlled cortical impact in mice. J Cereb Blood Flow Metab. 2011;31(1):351–61.CrossRefPubMed
19.
go back to reference Washington P, et al. Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-Beta in an Alzheimer's disease mouse model. J Neurotrauma. 2014;31(1):125–34.CrossRefPubMedPubMedCentral Washington P, et al. Experimental traumatic brain injury induces rapid aggregation and oligomerization of amyloid-Beta in an Alzheimer's disease mouse model. J Neurotrauma. 2014;31(1):125–34.CrossRefPubMedPubMedCentral
20.
go back to reference Loane D, et al. Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation. J Neuropathol Exp Neurol. 2014;73(1):14–29.CrossRefPubMedPubMedCentral Loane D, et al. Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation. J Neuropathol Exp Neurol. 2014;73(1):14–29.CrossRefPubMedPubMedCentral
21.
go back to reference Halliday MR, et al. Relationship between cyclophilin a levels and matrix metalloproteinase 9 activity in cerebrospinal fluid of cognitively normal apolipoprotein e4 carriers and blood-brain barrier breakdown. JAMA Neurol. 2013;70(9):1198–200.CrossRefPubMedPubMedCentral Halliday MR, et al. Relationship between cyclophilin a levels and matrix metalloproteinase 9 activity in cerebrospinal fluid of cognitively normal apolipoprotein e4 carriers and blood-brain barrier breakdown. JAMA Neurol. 2013;70(9):1198–200.CrossRefPubMedPubMedCentral
22.
go back to reference Halliday MR, et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J Cereb Blood Flow Metab. 2016;36(1):216–27.CrossRefPubMedPubMedCentral Halliday MR, et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J Cereb Blood Flow Metab. 2016;36(1):216–27.CrossRefPubMedPubMedCentral
23.
go back to reference Villapol S, Loane DJ, Burns MP. Sexual dimorphism in the inflammatory response to traumatic brain injury. Glia. 2017;65(9):1423–38.CrossRefPubMed Villapol S, Loane DJ, Burns MP. Sexual dimorphism in the inflammatory response to traumatic brain injury. Glia. 2017;65(9):1423–38.CrossRefPubMed
24.
go back to reference Sullivan PG, et al. Dose-response curve and optimal dosing regimen of cyclosporin a after traumatic brain injury in rats. Neuroscience. 2000;101(2):289–95.CrossRefPubMed Sullivan PG, et al. Dose-response curve and optimal dosing regimen of cyclosporin a after traumatic brain injury in rats. Neuroscience. 2000;101(2):289–95.CrossRefPubMed
25.
go back to reference Fe Lanfranco M, et al. Combination of fluorescent in situ hybridization (FISH) and immunofluorescence imaging for detection of cytokine expression in microglia/macrophage cells. Bio Protoc. 2017;7(22) Fe Lanfranco M, et al. Combination of fluorescent in situ hybridization (FISH) and immunofluorescence imaging for detection of cytokine expression in microglia/macrophage cells. Bio Protoc. 2017;7(22)
26.
go back to reference Yamakawa H, et al. Normalization of endothelial and inducible nitric oxide synthase expression in brain microvessels of spontaneously hypertensive rats by angiotensin II AT1 receptor inhibition. J Cereb Blood Flow Metab. 2003;23(3):371–80.CrossRefPubMed Yamakawa H, et al. Normalization of endothelial and inducible nitric oxide synthase expression in brain microvessels of spontaneously hypertensive rats by angiotensin II AT1 receptor inhibition. J Cereb Blood Flow Metab. 2003;23(3):371–80.CrossRefPubMed
27.
go back to reference Main B, et al. Type-1 interferons contribute to the neuroinflammatory response and disease progression of the MPTP mouse model of Parkinson's disease. Glia. 2016;64(9):1590–604.CrossRefPubMed Main B, et al. Type-1 interferons contribute to the neuroinflammatory response and disease progression of the MPTP mouse model of Parkinson's disease. Glia. 2016;64(9):1590–604.CrossRefPubMed
28.
go back to reference Main BS, et al. Type-I interferons mediate the neuroinflammatory response and neurotoxicity induced by rotenone. J Neurochem. 2017;141(1):75–85.CrossRefPubMed Main BS, et al. Type-I interferons mediate the neuroinflammatory response and neurotoxicity induced by rotenone. J Neurochem. 2017;141(1):75–85.CrossRefPubMed
29.
go back to reference Winer J, et al. Development and validation of real-time quantitative reverse transcriptase-polymerase chain reaction for monitoring gene expression in cardiac myocytes in vitro. Anal Biochem. 1999;270(1):41–9.CrossRefPubMed Winer J, et al. Development and validation of real-time quantitative reverse transcriptase-polymerase chain reaction for monitoring gene expression in cardiac myocytes in vitro. Anal Biochem. 1999;270(1):41–9.CrossRefPubMed
30.
31.
32.
go back to reference Bell RD, et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 2010;68(3):409–27.CrossRefPubMedPubMedCentral Bell RD, et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron. 2010;68(3):409–27.CrossRefPubMedPubMedCentral
33.
go back to reference Rodriguez-Baeza A, et al. Morphological features in human cortical brain microvessels after head injury: a three-dimensional and immunocytochemical study. Anat Rec A Discov Mol Cell Evol Biol. 2003;273((1):583–93.CrossRef Rodriguez-Baeza A, et al. Morphological features in human cortical brain microvessels after head injury: a three-dimensional and immunocytochemical study. Anat Rec A Discov Mol Cell Evol Biol. 2003;273((1):583–93.CrossRef
34.
go back to reference Kenney, K., et al., Cerebral vascular injury in traumatic brain injury. Exp Neurol, 2016 275 Pt 3: p. 353–366. Kenney, K., et al., Cerebral vascular injury in traumatic brain injury. Exp Neurol, 2016 275 Pt 3: p. 353–366.
35.
go back to reference Lin Y, et al. Blood-brain barrier permeability is positively correlated with cerebral microvascular perfusion in the early fluid percussion-injured brain of the rat. Lab Investig. 2012;92(11):1623–34.CrossRefPubMed Lin Y, et al. Blood-brain barrier permeability is positively correlated with cerebral microvascular perfusion in the early fluid percussion-injured brain of the rat. Lab Investig. 2012;92(11):1623–34.CrossRefPubMed
36.
go back to reference Park E, et al. An analysis of regional microvascular loss and recovery following two grades of fluid percussion trauma: a role for hypoxia-inducible factors in traumatic brain injury. J Cereb Blood Flow Metab. 2009;29(3):575–84.CrossRefPubMed Park E, et al. An analysis of regional microvascular loss and recovery following two grades of fluid percussion trauma: a role for hypoxia-inducible factors in traumatic brain injury. J Cereb Blood Flow Metab. 2009;29(3):575–84.CrossRefPubMed
37.
go back to reference Dixon CE, Lighthall JW, Anderson TE. Physiologic, histopathologic, and cineradiographic characterization of a new fluid-percussion model of experimental brain injury in the rat. J Neurotrauma. 1988;5(2):91–104.CrossRefPubMed Dixon CE, Lighthall JW, Anderson TE. Physiologic, histopathologic, and cineradiographic characterization of a new fluid-percussion model of experimental brain injury in the rat. J Neurotrauma. 1988;5(2):91–104.CrossRefPubMed
38.
go back to reference Dietrich WD, Alonso O, Halley M. Early microvascular and neuronal consequences of traumatic brain injury: a light and electron microscopic study in rats. J Neurotrauma. 1994;11(3):289–301.CrossRefPubMed Dietrich WD, Alonso O, Halley M. Early microvascular and neuronal consequences of traumatic brain injury: a light and electron microscopic study in rats. J Neurotrauma. 1994;11(3):289–301.CrossRefPubMed
39.
go back to reference Glushakova OY, Johnson D, Hayes RL. Delayed increases in microvascular pathology after experimental traumatic brain injury are associated with prolonged inflammation, blood-brain barrier disruption, and progressive white matter damage. J Neurotrauma. 2014;31(13):1180–93.CrossRefPubMed Glushakova OY, Johnson D, Hayes RL. Delayed increases in microvascular pathology after experimental traumatic brain injury are associated with prolonged inflammation, blood-brain barrier disruption, and progressive white matter damage. J Neurotrauma. 2014;31(13):1180–93.CrossRefPubMed
40.
go back to reference Li S, et al. Enoxaparin ameliorates post-traumatic brain injury edema and neurologic recovery, reducing cerebral leukocyte endothelial interactions and vessel permeability in vivo. J Trauma Acute Care Surg. 2015;79(1):78–84.CrossRefPubMedPubMedCentral Li S, et al. Enoxaparin ameliorates post-traumatic brain injury edema and neurologic recovery, reducing cerebral leukocyte endothelial interactions and vessel permeability in vivo. J Trauma Acute Care Surg. 2015;79(1):78–84.CrossRefPubMedPubMedCentral
41.
go back to reference Villapol S, Byrnes KR, Symes AJ. Temporal dynamics of cerebral blood flow, cortical damage, apoptosis, astrocyte-vasculature interaction and astrogliosis in the pericontusional region after traumatic brain injury. Front Neurol. 2014;5:82.CrossRefPubMedPubMedCentral Villapol S, Byrnes KR, Symes AJ. Temporal dynamics of cerebral blood flow, cortical damage, apoptosis, astrocyte-vasculature interaction and astrogliosis in the pericontusional region after traumatic brain injury. Front Neurol. 2014;5:82.CrossRefPubMedPubMedCentral
42.
go back to reference Cherian L, Goodman JC, Robertson C. Improved cerebrovascular function and reduced histological damage with darbepoietin alfa administration after cortical impact injury in rats. J Pharmacol Exp Ther. 2011;337(2):451–6.CrossRefPubMedPubMedCentral Cherian L, Goodman JC, Robertson C. Improved cerebrovascular function and reduced histological damage with darbepoietin alfa administration after cortical impact injury in rats. J Pharmacol Exp Ther. 2011;337(2):451–6.CrossRefPubMedPubMedCentral
43.
go back to reference Sangiorgi S, et al. Early-stage microvascular alterations of a new model of controlled cortical traumatic brain injury: 3D morphological analysis using scanning electron microscopy and corrosion casting. J Neurosurg. 2013;118(4):763–74.CrossRefPubMed Sangiorgi S, et al. Early-stage microvascular alterations of a new model of controlled cortical traumatic brain injury: 3D morphological analysis using scanning electron microscopy and corrosion casting. J Neurosurg. 2013;118(4):763–74.CrossRefPubMed
44.
45.
go back to reference Ho KM, et al. Prognostic significance of blood-brain barrier disruption in patients with severe nonpenetrating traumatic brain injury requiring decompressive craniectomy. J Neurosurg. 2014;121(3):674–9.CrossRefPubMed Ho KM, et al. Prognostic significance of blood-brain barrier disruption in patients with severe nonpenetrating traumatic brain injury requiring decompressive craniectomy. J Neurosurg. 2014;121(3):674–9.CrossRefPubMed
46.
go back to reference Saw MM, et al. Differential disruption of blood-brain barrier in severe traumatic brain injury. Neurocrit Care. 2014;20(2):209–16.CrossRefPubMed Saw MM, et al. Differential disruption of blood-brain barrier in severe traumatic brain injury. Neurocrit Care. 2014;20(2):209–16.CrossRefPubMed
47.
go back to reference Stahel PF, et al. Intrathecal levels of complement-derived soluble membrane attack complex (sC5b-9) correlate with blood-brain barrier dysfunction in patients with traumatic brain injury. J Neurotrauma. 2001;18(8):773–81.CrossRefPubMed Stahel PF, et al. Intrathecal levels of complement-derived soluble membrane attack complex (sC5b-9) correlate with blood-brain barrier dysfunction in patients with traumatic brain injury. J Neurotrauma. 2001;18(8):773–81.CrossRefPubMed
48.
go back to reference Csuka E, et al. 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. 1999;101(2):211–21.CrossRefPubMed Csuka E, et al. 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. 1999;101(2):211–21.CrossRefPubMed
49.
go back to reference Tomkins O, et al. Blood-brain barrier breakdown following traumatic brain injury: a possible role in posttraumatic epilepsy. Cardiovasc Psychiatry Neurol. 2011;2011:765923.CrossRefPubMedPubMedCentral Tomkins O, et al. Blood-brain barrier breakdown following traumatic brain injury: a possible role in posttraumatic epilepsy. Cardiovasc Psychiatry Neurol. 2011;2011:765923.CrossRefPubMedPubMedCentral
50.
go back to reference Hay JR, et al. Blood-brain barrier disruption is an early event that may persist for many years after traumatic brain injury in humans. J Neuropathol Exp Neurol. 2015;74(12):1147–57.PubMed Hay JR, et al. Blood-brain barrier disruption is an early event that may persist for many years after traumatic brain injury in humans. J Neuropathol Exp Neurol. 2015;74(12):1147–57.PubMed
51.
go back to reference Barzo P, et al. Magnetic resonance imaging-monitored acute blood-brain barrier changes in experimental traumatic brain injury. J Neurosurg. 1996;85(6):1113–21.CrossRefPubMed Barzo P, et al. Magnetic resonance imaging-monitored acute blood-brain barrier changes in experimental traumatic brain injury. J Neurosurg. 1996;85(6):1113–21.CrossRefPubMed
52.
go back to reference Beaumont A, et al. 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.CrossRefPubMed Beaumont A, et al. 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.CrossRefPubMed
53.
go back to reference Higashida T, et al. The role of hypoxia-inducible factor-1alpha, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2011;114(1):92–101.CrossRefPubMed Higashida T, et al. The role of hypoxia-inducible factor-1alpha, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg. 2011;114(1):92–101.CrossRefPubMed
54.
go back to reference Shapira Y, et al. Blood-brain barrier permeability, cerebral edema, and neurologic function after closed head injury in rats. Anesth Analg. 1993;77(1):141–8.CrossRefPubMed Shapira Y, et al. Blood-brain barrier permeability, cerebral edema, and neurologic function after closed head injury in rats. Anesth Analg. 1993;77(1):141–8.CrossRefPubMed
55.
go back to reference Baldwin SA, et al. Blood-brain barrier breach following cortical contusion in the rat. J Neurosurg. 1996;85(3):476–81.CrossRefPubMed Baldwin SA, et al. Blood-brain barrier breach following cortical contusion in the rat. J Neurosurg. 1996;85(3):476–81.CrossRefPubMed
56.
go back to reference Si D, et al. Progesterone protects blood-brain barrier function and improves neurological outcome following traumatic brain injury in rats. Exp Ther Med. 2014;8(3):1010–4.CrossRefPubMedPubMedCentral Si D, et al. Progesterone protects blood-brain barrier function and improves neurological outcome following traumatic brain injury in rats. Exp Ther Med. 2014;8(3):1010–4.CrossRefPubMedPubMedCentral
57.
go back to reference Baskaya MK, et al. The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett. 1997;226(1):33–6.CrossRefPubMed Baskaya MK, et al. The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett. 1997;226(1):33–6.CrossRefPubMed
58.
go back to reference Yuan F, et al. SIRT2 inhibition exacerbates neuroinflammation and blood-brain barrier disruption in experimental traumatic brain injury by enhancing NF-kappaB p65 acetylation and activation. J Neurochem. 2016;136(3):581–93.CrossRefPubMed Yuan F, et al. SIRT2 inhibition exacerbates neuroinflammation and blood-brain barrier disruption in experimental traumatic brain injury by enhancing NF-kappaB p65 acetylation and activation. J Neurochem. 2016;136(3):581–93.CrossRefPubMed
59.
go back to reference Li H, et al. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016;1642:70–8.CrossRefPubMed Li H, et al. Sodium butyrate exerts neuroprotective effects by restoring the blood-brain barrier in traumatic brain injury mice. Brain Res. 2016;1642:70–8.CrossRefPubMed
60.
go back to reference Tao X, et al. Protective actions of PJ34, a poly(ADP-ribose)polymerase inhibitor, on the blood-brain barrier after traumatic brain injury in mice. Neuroscience. 2015;291:26–36.CrossRefPubMed Tao X, et al. Protective actions of PJ34, a poly(ADP-ribose)polymerase inhibitor, on the blood-brain barrier after traumatic brain injury in mice. Neuroscience. 2015;291:26–36.CrossRefPubMed
61.
go back to reference Dash PK, et al. Activation of alpha 7 cholinergic nicotinic receptors reduce blood-brain barrier permeability following experimental traumatic brain injury. J Neurosci. 2016;36(9):2809–18.CrossRefPubMedPubMedCentral Dash PK, et al. Activation of alpha 7 cholinergic nicotinic receptors reduce blood-brain barrier permeability following experimental traumatic brain injury. J Neurosci. 2016;36(9):2809–18.CrossRefPubMedPubMedCentral
62.
go back to reference Yao X, et al. Mildly reduced brain swelling and improved neurological outcome in Aquaporin-4 knockout mice following controlled cortical impact brain injury. J Neurotrauma. 2015;32(19):1458–64.CrossRefPubMedPubMedCentral Yao X, et al. Mildly reduced brain swelling and improved neurological outcome in Aquaporin-4 knockout mice following controlled cortical impact brain injury. J Neurotrauma. 2015;32(19):1458–64.CrossRefPubMedPubMedCentral
63.
go back to reference Chen CC, et al. Wogonin improves histological and functional outcomes, and reduces activation of TLR4/NF-kappaB signaling after experimental traumatic brain injury. PLoS One. 2012;7(1):e30294.CrossRefPubMedPubMedCentral Chen CC, et al. Wogonin improves histological and functional outcomes, and reduces activation of TLR4/NF-kappaB signaling after experimental traumatic brain injury. PLoS One. 2012;7(1):e30294.CrossRefPubMedPubMedCentral
64.
go back to reference Price L, Wilson C, Grant G. Chapter 4: Blood-brain barrier pathophysiology following traumatic brain injury. In: Laskowitz D, Grant G, editors. Translational Research in Traumatic Brain Injury. Boca Raton: CRC Press/Taylor and Francis Group; 2016. Price L, Wilson C, Grant G. Chapter 4: Blood-brain barrier pathophysiology following traumatic brain injury. In: Laskowitz D, Grant G, editors. Translational Research in Traumatic Brain Injury. Boca Raton: CRC Press/Taylor and Francis Group; 2016.
65.
66.
go back to reference Meng Q, et al. Traumatic brain injury induces genome-wide transcriptomic, Methylomic, and network perturbations in brain and blood predicting neurological disorders. EBioMedicine. 2017;16:184–94.CrossRefPubMedPubMedCentral Meng Q, et al. Traumatic brain injury induces genome-wide transcriptomic, Methylomic, and network perturbations in brain and blood predicting neurological disorders. EBioMedicine. 2017;16:184–94.CrossRefPubMedPubMedCentral
67.
go back to reference Cartagena CM, et al. Cortical injury increases cholesterol 24S hydroxylase (Cyp46) levels in the rat brain. JNeurotrauma. 2008;25(9):1087–98.CrossRef Cartagena CM, et al. Cortical injury increases cholesterol 24S hydroxylase (Cyp46) levels in the rat brain. JNeurotrauma. 2008;25(9):1087–98.CrossRef
68.
go back to reference Loane DJ, et al. Modulation of ABCA1 by an LXR agonist reduces beta-amyloid levels and improves outcome after traumatic brain injury. J Neurotrauma. 2011;28(2):225–36.CrossRefPubMedPubMedCentral Loane DJ, et al. Modulation of ABCA1 by an LXR agonist reduces beta-amyloid levels and improves outcome after traumatic brain injury. J Neurotrauma. 2011;28(2):225–36.CrossRefPubMedPubMedCentral
69.
go back to reference Xu Q, et al. Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci. 2006;26(19):4985–94.CrossRefPubMed Xu Q, et al. Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci. 2006;26(19):4985–94.CrossRefPubMed
70.
go back to reference Wilhelmus MM, et al. Apolipoprotein E genotype regulates amyloid-beta cytotoxicity. J Neurosci. 2005;25(14):3621–7.CrossRefPubMed Wilhelmus MM, et al. Apolipoprotein E genotype regulates amyloid-beta cytotoxicity. J Neurosci. 2005;25(14):3621–7.CrossRefPubMed
71.
go back to reference Casey CS, et al. Apolipoprotein E inhibits cerebrovascular Pericyte mobility through a RhoA protein-mediated pathway. J Biol Chem. 2015;290(22):14208–17.CrossRefPubMedPubMedCentral Casey CS, et al. Apolipoprotein E inhibits cerebrovascular Pericyte mobility through a RhoA protein-mediated pathway. J Biol Chem. 2015;290(22):14208–17.CrossRefPubMedPubMedCentral
72.
go back to reference Enerson BE, Drewes LR. The rat blood-brain barrier transcriptome. J Cereb Blood Flow Metab. 2006;26(7):959–73.CrossRefPubMed Enerson BE, Drewes LR. The rat blood-brain barrier transcriptome. J Cereb Blood Flow Metab. 2006;26(7):959–73.CrossRefPubMed
73.
go back to reference Zozulya A, Weidenfeller C, Galla HJ. Pericyte-endothelial cell interaction increases MMP-9 secretion at the blood-brain barrier in vitro. Brain Res. 2008;1189:1–11.CrossRefPubMed Zozulya A, Weidenfeller C, Galla HJ. Pericyte-endothelial cell interaction increases MMP-9 secretion at the blood-brain barrier in vitro. Brain Res. 2008;1189:1–11.CrossRefPubMed
74.
go back to reference Storck SE, et al. Endothelial LRP1 transports amyloid-beta(1-42) across the blood-brain barrier. J Clin Invest. 2016;126(1):123–36.CrossRefPubMed Storck SE, et al. Endothelial LRP1 transports amyloid-beta(1-42) across the blood-brain barrier. J Clin Invest. 2016;126(1):123–36.CrossRefPubMed
75.
go back to reference Alata W, et al. Human apolipoprotein E varepsilon4 expression impairs cerebral vascularization and blood-brain barrier function in mice. J Cereb Blood Flow Metab. 2015;35(1):86–94.CrossRefPubMed Alata W, et al. Human apolipoprotein E varepsilon4 expression impairs cerebral vascularization and blood-brain barrier function in mice. J Cereb Blood Flow Metab. 2015;35(1):86–94.CrossRefPubMed
76.
77.
go back to reference Winkler EA, et al. Blood-spinal cord barrier pericyte reductions contribute to increased capillary permeability. J Cereb Blood Flow Metab. 2012;32(10):1841–52.CrossRefPubMedPubMedCentral Winkler EA, et al. Blood-spinal cord barrier pericyte reductions contribute to increased capillary permeability. J Cereb Blood Flow Metab. 2012;32(10):1841–52.CrossRefPubMedPubMedCentral
78.
go back to reference Zehendner CM, et al. Traumatic brain injury results in rapid pericyte loss followed by reactive pericytosis in the cerebral cortex. Sci Rep. 2015;5:13497.CrossRefPubMedPubMedCentral Zehendner CM, et al. Traumatic brain injury results in rapid pericyte loss followed by reactive pericytosis in the cerebral cortex. Sci Rep. 2015;5:13497.CrossRefPubMedPubMedCentral
79.
go back to reference Dore-Duffy P, et al. Pericyte migration from the vascular wall in response to traumatic brain injury. Microvasc Res. 2000;60(1):55–69.CrossRefPubMed Dore-Duffy P, et al. Pericyte migration from the vascular wall in response to traumatic brain injury. Microvasc Res. 2000;60(1):55–69.CrossRefPubMed
80.
go back to reference Takata F, et al. Brain pericytes among cells constituting the blood-brain barrier are highly sensitive to tumor necrosis factor-alpha, releasing matrix metalloproteinase-9 and migrating in vitro. J Neuroinflammation. 2011;8:106.CrossRefPubMedPubMedCentral Takata F, et al. Brain pericytes among cells constituting the blood-brain barrier are highly sensitive to tumor necrosis factor-alpha, releasing matrix metalloproteinase-9 and migrating in vitro. J Neuroinflammation. 2011;8:106.CrossRefPubMedPubMedCentral
81.
go back to reference Vilalta A, et al. Moderate and severe traumatic brain injury induce early overexpression of systemic and brain gelatinases. Intensive Care Med. 2008;34(8):1384–92.CrossRefPubMed Vilalta A, et al. Moderate and severe traumatic brain injury induce early overexpression of systemic and brain gelatinases. Intensive Care Med. 2008;34(8):1384–92.CrossRefPubMed
82.
go back to reference Suehiro E, 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.CrossRefPubMed Suehiro E, 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.CrossRefPubMed
83.
go back to reference Tsuji D, et al. Specific induction of macrophage inflammatory protein 1-alpha in glial cells of Sandhoff disease model mice associated with accumulation of N-acetylhexosaminyl glycoconjugates. J Neurochem. 2005;92(6):1497–507.CrossRefPubMed Tsuji D, et al. Specific induction of macrophage inflammatory protein 1-alpha in glial cells of Sandhoff disease model mice associated with accumulation of N-acetylhexosaminyl glycoconjugates. J Neurochem. 2005;92(6):1497–507.CrossRefPubMed
84.
go back to reference Zhang H, et al. N-Acetylglucosamine 6-O-sulfotransferase-1 is required for brain keratan sulfate biosynthesis and glial scar formation after brain injury. Glycobiology. 2006;16(8):702–10.CrossRefPubMed Zhang H, et al. N-Acetylglucosamine 6-O-sulfotransferase-1 is required for brain keratan sulfate biosynthesis and glial scar formation after brain injury. Glycobiology. 2006;16(8):702–10.CrossRefPubMed
85.
go back to reference Zhang H, Uchimura K, Kadomatsu K. Brain keratan sulfate and glial scar formation. Ann N Y Acad Sci. 2006;1086:81–90.CrossRefPubMed Zhang H, Uchimura K, Kadomatsu K. Brain keratan sulfate and glial scar formation. Ann N Y Acad Sci. 2006;1086:81–90.CrossRefPubMed
86.
go back to reference Colton CA, et al. Lectin staining of cultured CNS microglia. J Histochem Cytochem. 1992;40(4):505–12.CrossRefPubMed Colton CA, et al. Lectin staining of cultured CNS microglia. J Histochem Cytochem. 1992;40(4):505–12.CrossRefPubMed
87.
go back to reference Kelley BJ, Lifshitz J, Povlishock JT. Neuroinflammatory responses after experimental diffuse traumatic brain injury. J Neuropathol Exp Neurol. 2007;66(11):989–1001.CrossRefPubMed Kelley BJ, Lifshitz J, Povlishock JT. Neuroinflammatory responses after experimental diffuse traumatic brain injury. J Neuropathol Exp Neurol. 2007;66(11):989–1001.CrossRefPubMed
Metadata
Title
Apolipoprotein E4 impairs spontaneous blood brain barrier repair following traumatic brain injury
Authors
Bevan S. Main
Sonia Villapol
Stephanie S. Sloley
David J. Barton
Maia Parsadanian
Chinyere Agbaegbu
Kathryn Stefos
Mondona S. McCann
Patricia M. Washington
Olga C. Rodriguez
Mark P. Burns
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Molecular Neurodegeneration / Issue 1/2018
Electronic ISSN: 1750-1326
DOI
https://doi.org/10.1186/s13024-018-0249-5

Other articles of this Issue 1/2018

Molecular Neurodegeneration 1/2018 Go to the issue