Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2015

Open Access 01-12-2015 | Research

Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit

Authors: William A. Banks, Alicia M. Gray, Michelle A. Erickson, Therese S. Salameh, Mamatha Damodarasamy, Nader Sheibani, James S. Meabon, Emily E. Wing, Yoichi Morofuji, David G. Cook, May J. Reed

Published in: Journal of Neuroinflammation | Issue 1/2015

Login to get access

Abstract

Background

Disruption of the blood-brain barrier (BBB) occurs in many diseases and is often mediated by inflammatory and neuroimmune mechanisms. Inflammation is well established as a cause of BBB disruption, but many mechanistic questions remain.

Methods

We used lipopolysaccharide (LPS) to induce inflammation and BBB disruption in mice. BBB disruption was measured using 14C-sucrose and radioactively labeled albumin. Brain cytokine responses were measured using multiplex technology and dependence on cyclooxygenase (COX) and oxidative stress determined by treatments with indomethacin and N-acetylcysteine. Astrocyte and microglia/macrophage responses were measured using brain immunohistochemistry. In vitro studies used Transwell cultures of primary brain endothelial cells co- or tri-cultured with astrocytes and pericytes to measure effects of LPS on transendothelial electrical resistance (TEER), cellular distribution of tight junction proteins, and permeability to 14C-sucrose and radioactive albumin.

Results

In comparison to LPS-induced weight loss, the BBB was relatively resistant to LPS-induced disruption. Disruption occurred only with the highest dose of LPS and was most evident in the frontal cortex, thalamus, pons-medulla, and cerebellum with no disruption in the hypothalamus. The in vitro and in vivo patterns of LPS-induced disruption as measured with 14C-sucrose, radioactive albumin, and TEER suggested involvement of both paracellular and transcytotic pathways. Disruption as measured with albumin and 14C-sucrose, but not TEER, was blocked by indomethacin. N-acetylcysteine did not affect disruption. In vivo, the measures of neuroinflammation induced by LPS were mainly not reversed by indomethacin. In vitro, the effects on LPS and indomethacin were not altered when brain endothelial cells (BECs) were cultured with astrocytes or pericytes.

Conclusions

The BBB is relatively resistant to LPS-induced disruption with some brain regions more vulnerable than others. LPS-induced disruption appears is to be dependent on COX but not on oxidative stress. Based on in vivo and in vitro measures of neuroinflammation, it appears that astrocytes, microglia/macrophages, and pericytes play little role in the LPS-mediated disruption of the BBB.
Literature
1.
go back to reference Skoog T. On the vital staining of the central nervous system. Acta Otolaryngologica. 1937;25:365–78.CrossRef Skoog T. On the vital staining of the central nervous system. Acta Otolaryngologica. 1937;25:365–78.CrossRef
2.
go back to reference Rapoport SI. Pathological alterations of the blood-brain barrier. In: Blood-brain barrier in physiology and medicine. New York: Raven; 1976. p. 129–52. Rapoport SI. Pathological alterations of the blood-brain barrier. In: Blood-brain barrier in physiology and medicine. New York: Raven; 1976. p. 129–52.
3.
go back to reference Allen IV. The effect of bacterial pyrogen on the blood-brain barrier for trypan blue. J Pathol Bacteriol. 1965;89:481–94.CrossRefPubMed Allen IV. The effect of bacterial pyrogen on the blood-brain barrier for trypan blue. J Pathol Bacteriol. 1965;89:481–94.CrossRefPubMed
4.
go back to reference Eckman PL, King WM, Brunson JG. Studies on the blood brain barrier. I. Effects produced by a single injection of gram-negative endotoxin on the permeability of the cerebral vessels. Am J Pathol. 1958;34:631–43.PubMedCentralPubMed Eckman PL, King WM, Brunson JG. Studies on the blood brain barrier. I. Effects produced by a single injection of gram-negative endotoxin on the permeability of the cerebral vessels. Am J Pathol. 1958;34:631–43.PubMedCentralPubMed
5.
go back to reference Persidsky Y, Stins M, Way D, Witte MH, Weinand M, Kim KS, et al. A model for monocyte migration through the blood-brain barrier during HIV-1 encephalitis. J Immunol. 1997;158:3499–510.PubMed Persidsky Y, Stins M, Way D, Witte MH, Weinand M, Kim KS, et al. A model for monocyte migration through the blood-brain barrier during HIV-1 encephalitis. J Immunol. 1997;158:3499–510.PubMed
6.
go back to reference Banks WA, Kastin AJ, Brennan JM, Vallance KL. Adsorptive endocytosis of HIV-1gp120 by blood-brain barrier is enhanced by lipopolysaccharide. Exp Neurol. 1999;156:165–71.CrossRefPubMed Banks WA, Kastin AJ, Brennan JM, Vallance KL. Adsorptive endocytosis of HIV-1gp120 by blood-brain barrier is enhanced by lipopolysaccharide. Exp Neurol. 1999;156:165–71.CrossRefPubMed
7.
go back to reference Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001;896:36–42.CrossRefPubMed Xaio H, Banks WA, Niehoff ML, Morley JE. Effect of LPS on the permeability of the blood-brain barrier to insulin. Brain Res. 2001;896:36–42.CrossRefPubMed
8.
go back to reference Minami T, Okazaki J, Kawabata A, Kuroda R, Okazaki Y. Penetration of cisplatin into mouse brain by lipopolysaccharide. Toxicology. 1998;130:107–13.CrossRefPubMed Minami T, Okazaki J, Kawabata A, Kuroda R, Okazaki Y. Penetration of cisplatin into mouse brain by lipopolysaccharide. Toxicology. 1998;130:107–13.CrossRefPubMed
9.
go back to reference Pan W, Yu C, Hsuchou H, Zhang Y, Kastin AJ. Neuroinflammation facilitates LIF entry into brain: role of TNF. Am J Physiol Cell Physiol. 2008;294:C1436–42.PubMedCentralCrossRefPubMed Pan W, Yu C, Hsuchou H, Zhang Y, Kastin AJ. Neuroinflammation facilitates LIF entry into brain: role of TNF. Am J Physiol Cell Physiol. 2008;294:C1436–42.PubMedCentralCrossRefPubMed
10.
go back to reference Tucsek Z, Toth P, Sosnowska D, Gautam T, Mitschelen M, Koller A, et al. Obesity in aging exacerbates blood-brain barrier disruption, neuroinflammation, and oxidative stress in the mouse hippocampus: effects on expression of genes involved in beta-amyloid generation and Alzheimer’s disease. J Gerontol A Biol Sci Med Sci. 2013, epub. Tucsek Z, Toth P, Sosnowska D, Gautam T, Mitschelen M, Koller A, et al. Obesity in aging exacerbates blood-brain barrier disruption, neuroinflammation, and oxidative stress in the mouse hippocampus: effects on expression of genes involved in beta-amyloid generation and Alzheimer’s disease. J Gerontol A Biol Sci Med Sci. 2013, epub.
11.
go back to reference Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease. Brain Pathol. 2012;23:303–10.PubMedCentralCrossRefPubMed Sengillo JD, Winkler EA, Walker CT, Sullivan JS, Johnson M, Zlokovic BV. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer’s disease. Brain Pathol. 2012;23:303–10.PubMedCentralCrossRefPubMed
12.
go back to reference Starr JM, Wardlaw J, Ferguson K, MacLullich A, Deary IJ, Marshall I. Increased blood-brain barrier permeability in type II diabetes demonstrated by gadolinium magnetic resonance imaging. J Neurol Neurosurg Psychiatry. 2003;74:70–6.PubMedCentralCrossRefPubMed Starr JM, Wardlaw J, Ferguson K, MacLullich A, Deary IJ, Marshall I. Increased blood-brain barrier permeability in type II diabetes demonstrated by gadolinium magnetic resonance imaging. J Neurol Neurosurg Psychiatry. 2003;74:70–6.PubMedCentralCrossRefPubMed
13.
go back to reference Abraham CS, Harada N, Deli MA, Niwa M. Transient forebrain ischemia increases the blood-brain barrier permeability for albumin in stroke-prone spontaneously hypertensive rats. Cell Mol Neurobiol. 2003;22:455–62.CrossRef Abraham CS, Harada N, Deli MA, Niwa M. Transient forebrain ischemia increases the blood-brain barrier permeability for albumin in stroke-prone spontaneously hypertensive rats. Cell Mol Neurobiol. 2003;22:455–62.CrossRef
14.
go back to reference Pozzilli C, Bernardi S, Mansi L, Picozzi P, Iannotti F, Alfano B, et al. Quantitative assessment of the blood-brain barrier permeability in multiple sclerosis using 68-Ga-EDTA and positron emission tomography. J Neurol Neurosurg Psychiatry. 1988;51:1058–62.PubMedCentralCrossRefPubMed Pozzilli C, Bernardi S, Mansi L, Picozzi P, Iannotti F, Alfano B, et al. Quantitative assessment of the blood-brain barrier permeability in multiple sclerosis using 68-Ga-EDTA and positron emission tomography. J Neurol Neurosurg Psychiatry. 1988;51:1058–62.PubMedCentralCrossRefPubMed
17.
go back to reference Mayhan WG, Heistad DD. Permeability of blood-brain barrier to various sized molecules. Am J Physiology. 1985;248:H712–8. Mayhan WG, Heistad DD. Permeability of blood-brain barrier to various sized molecules. Am J Physiology. 1985;248:H712–8.
18.
go back to reference Ziylan YZ, Robinson PJ, Rapoport SI. Blood-brain barrier permeability to sucrose and dextran after osmotic opening. Am J Physiol. 1984;247:R634–8.PubMed Ziylan YZ, Robinson PJ, Rapoport SI. Blood-brain barrier permeability to sucrose and dextran after osmotic opening. Am J Physiol. 1984;247:R634–8.PubMed
19.
go back to reference Knowland D, Arac A, Sekiguchi KJ, Hsu M, Lutz SE, Perrino J, et al. Stepwise recruitment of transcellular and paracellular pathways underlies blood-brain barrier breakdown in stroke. Neuron. 2014;82:603–17.PubMedCentralCrossRefPubMed Knowland D, Arac A, Sekiguchi KJ, Hsu M, Lutz SE, Perrino J, et al. Stepwise recruitment of transcellular and paracellular pathways underlies blood-brain barrier breakdown in stroke. Neuron. 2014;82:603–17.PubMedCentralCrossRefPubMed
20.
go back to reference Fleegal-DeMotta MA, Dohgu S, Banks WA. Angiotensin II modulates BBB permeability via activation of the AT1 receptor in brain endothelial cells. J Cereb Blood Flow Metab. 2009;29:640–7.CrossRefPubMed Fleegal-DeMotta MA, Dohgu S, Banks WA. Angiotensin II modulates BBB permeability via activation of the AT1 receptor in brain endothelial cells. J Cereb Blood Flow Metab. 2009;29:640–7.CrossRefPubMed
21.
go back to reference De Vries HE, Blom-Roosemalen MCM, De Boer AG, van Berkel TJ, Breimer DD, Kuiper J. Effect of endotoxin on permeability of bovine cerebral endothelial cell layers in vitro. J Pharmacol Exp Ther. 1996;277:1418–23.PubMed De Vries HE, Blom-Roosemalen MCM, De Boer AG, van Berkel TJ, Breimer DD, Kuiper J. Effect of endotoxin on permeability of bovine cerebral endothelial cell layers in vitro. J Pharmacol Exp Ther. 1996;277:1418–23.PubMed
22.
go back to reference Minami T, Okazaki J, Kawabata A, Kawaki H, Okazaki Y, Tohno Y. Roles of nitric oxide and prostaglandins in the increased permeability of the blood-brain barrier caused by lipopolysaccharide. Environ Toxicol Pharmacol. 1998;5:35–41.CrossRefPubMed Minami T, Okazaki J, Kawabata A, Kawaki H, Okazaki Y, Tohno Y. Roles of nitric oxide and prostaglandins in the increased permeability of the blood-brain barrier caused by lipopolysaccharide. Environ Toxicol Pharmacol. 1998;5:35–41.CrossRefPubMed
23.
go back to reference Jaeger JB, Dohgu S, Lynch JL, Fleegal-DeMotta MA, Banks WA. Effects of lipopolysaccharide on the blood-brain barrier transport of amyloid beta protein: a mechanism for inflammation in the progression of Alzheimer’s disease. Brain Behav Immun. 2009;23:507–17.PubMedCentralCrossRefPubMed Jaeger JB, Dohgu S, Lynch JL, Fleegal-DeMotta MA, Banks WA. Effects of lipopolysaccharide on the blood-brain barrier transport of amyloid beta protein: a mechanism for inflammation in the progression of Alzheimer’s disease. Brain Behav Immun. 2009;23:507–17.PubMedCentralCrossRefPubMed
24.
go back to reference Salkeni MA, Lynch JL, Price TO, Banks WA. Lipopolysaccharide impairs blood-brain barrier P-glycoprotein function in mice through prostaglandin- and nitric oxide-independent pathways and nitric oxide-independent pathways. J Neuroimmune Pharmacology. 2009;4:276–82.CrossRef Salkeni MA, Lynch JL, Price TO, Banks WA. Lipopolysaccharide impairs blood-brain barrier P-glycoprotein function in mice through prostaglandin- and nitric oxide-independent pathways and nitric oxide-independent pathways. J Neuroimmune Pharmacology. 2009;4:276–82.CrossRef
25.
go back to reference Zhang X, Banerjee A, Banks WA, Ercal N. N-acetyl amide protects against methamphetamine-induced oxidative stress and neurotoxicity in immortalized human brain endothelial cells. Brain Res. 2009;1275:87–95.PubMedCentralCrossRefPubMed Zhang X, Banerjee A, Banks WA, Ercal N. N-acetyl amide protects against methamphetamine-induced oxidative stress and neurotoxicity in immortalized human brain endothelial cells. Brain Res. 2009;1275:87–95.PubMedCentralCrossRefPubMed
26.
go back to reference Manda KR, Banerjee A, Banks WA, Ercal N. Highly active antiretroviral therapy drug combination induces oxidative stress and mitochondrial dysfunction in immortalized human blood-brain barrier endothelial cells. Free Radic Biol Med. 2011;50:801–10.CrossRefPubMed Manda KR, Banerjee A, Banks WA, Ercal N. Highly active antiretroviral therapy drug combination induces oxidative stress and mitochondrial dysfunction in immortalized human blood-brain barrier endothelial cells. Free Radic Biol Med. 2011;50:801–10.CrossRefPubMed
27.
go back to reference Erickson MA, Hansen K, Banks WA. Inflammation-induced dysfunction of the low-density lipoprotein receptor-related protein-1 at the blood-brain barrier: protection by the antioxidant N-acetylcysteine. Brain Behav Immun. 2012;26:1085–94.PubMedCentralCrossRefPubMed Erickson MA, Hansen K, Banks WA. Inflammation-induced dysfunction of the low-density lipoprotein receptor-related protein-1 at the blood-brain barrier: protection by the antioxidant N-acetylcysteine. Brain Behav Immun. 2012;26:1085–94.PubMedCentralCrossRefPubMed
28.
go back to reference Coisne C, Dehouck L, Faveeuw C, Delplace Y, Miller F, Landry C, et al. Mouse syngenic in vitro blood-brain barrier model: a new tool to examine inflammatory events in cerebral endothelium. Lab Invest. 2005;85:734–46.CrossRefPubMed Coisne C, Dehouck L, Faveeuw C, Delplace Y, Miller F, Landry C, et al. Mouse syngenic in vitro blood-brain barrier model: a new tool to examine inflammatory events in cerebral endothelium. Lab Invest. 2005;85:734–46.CrossRefPubMed
29.
go back to reference Jacob A, Hack B, Chiang E, Garcia JG, Quigg RJ, Alexander JJ. C5a alters blood-brain barrier integrity in experimental lupus. FASEB J. 2010;24:1682–8.PubMedCentralCrossRefPubMed Jacob A, Hack B, Chiang E, Garcia JG, Quigg RJ, Alexander JJ. C5a alters blood-brain barrier integrity in experimental lupus. FASEB J. 2010;24:1682–8.PubMedCentralCrossRefPubMed
30.
go back to reference Perriere N, Demeuse P, Garcia E, Regina A, Debray M, Andreux JP, et al. Puromycin-based purification of rat brain capillary endothelial cell cultures. J Neurochem. 2005;93:279–89.CrossRefPubMed Perriere N, Demeuse P, Garcia E, Regina A, Debray M, Andreux JP, et al. Puromycin-based purification of rat brain capillary endothelial cell cultures. J Neurochem. 2005;93:279–89.CrossRefPubMed
31.
go back to reference Shah GN, Price TO, Banks WA, Morofuji Y, Kovac A, Ercal N, et al. Pharmacological inhibition of mitochondrial carbonic anhydrases protects mouse cerebral pericytes from high glucose-induced oxidative stress and apoptosis. J Pharmacol Exp Therap. 2013;344:637–45.CrossRef Shah GN, Price TO, Banks WA, Morofuji Y, Kovac A, Ercal N, et al. Pharmacological inhibition of mitochondrial carbonic anhydrases protects mouse cerebral pericytes from high glucose-induced oxidative stress and apoptosis. J Pharmacol Exp Therap. 2013;344:637–45.CrossRef
32.
go back to reference Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Kittel A, et al. A new blood-brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54:253–63.CrossRefPubMed Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Kittel A, et al. A new blood-brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int. 2009;54:253–63.CrossRefPubMed
33.
go back to reference Hoheisel D, Nits T, Franke H, Wegener J, Hakvoort A, Tilling T, et al. Hydrocortisone reinforces the blood-brain barrier properties in a serum free cell culture system. Biochem Biophys Res Commun. 1998;217:312–5.CrossRef Hoheisel D, Nits T, Franke H, Wegener J, Hakvoort A, Tilling T, et al. Hydrocortisone reinforces the blood-brain barrier properties in a serum free cell culture system. Biochem Biophys Res Commun. 1998;217:312–5.CrossRef
34.
go back to reference Dehouck MP, Jolliet-Riant P, Bree F, Fruchart JC, Cecchelli R, Tillement JP. Drug transfer across the blood-brain barrier: correlation between in vitro and in vivo models. J Neurochem. 1992;58:1790–7.CrossRefPubMed Dehouck MP, Jolliet-Riant P, Bree F, Fruchart JC, Cecchelli R, Tillement JP. Drug transfer across the blood-brain barrier: correlation between in vitro and in vivo models. J Neurochem. 1992;58:1790–7.CrossRefPubMed
35.
go back to reference Scimemi A, Meabon JS, Woltjer RL, Sullivan JM, Diamond JS, Cook DG. Amyloid-β1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1. J Neurosci. 2013;33:5312–8.PubMedCentralCrossRefPubMed Scimemi A, Meabon JS, Woltjer RL, Sullivan JM, Diamond JS, Cook DG. Amyloid-β1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1. J Neurosci. 2013;33:5312–8.PubMedCentralCrossRefPubMed
36.
go back to reference Daws MR, Lanier LL, Seaman WE, Ryan JC. Cloning and characterization of a novel mouse myeloid DAP 12-associated receptor family. Eur J Immunol. 2001;31:783–91.CrossRefPubMed Daws MR, Lanier LL, Seaman WE, Ryan JC. Cloning and characterization of a novel mouse myeloid DAP 12-associated receptor family. Eur J Immunol. 2001;31:783–91.CrossRefPubMed
37.
go back to reference Kleinberger G, Yamanishi Y, Suarez-Calvet M, Czirr E, Lohmann E, Cuyvers E, et al. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci Transl Med. 2014;6:243ra286.CrossRef Kleinberger G, Yamanishi Y, Suarez-Calvet M, Czirr E, Lohmann E, Cuyvers E, et al. TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis. Sci Transl Med. 2014;6:243ra286.CrossRef
38.
go back to reference Nonaka N, Shioda S, Banks WA. Effect of lipopolysaccharide on the transport of pituitary adenylate cyclase activating polypeptide across the blood-brain barrier. Exp Neurol. 2005;191:137–44.CrossRefPubMed Nonaka N, Shioda S, Banks WA. Effect of lipopolysaccharide on the transport of pituitary adenylate cyclase activating polypeptide across the blood-brain barrier. Exp Neurol. 2005;191:137–44.CrossRefPubMed
39.
go back to reference Erickson MA, Hartvigson PE, Morofuji Y, Owen JB, Butterfield DA, Banks WA. Lipopolysaccharide impairs amyloid beta efflux from brain: altered vascular sequestration, cerebrospinal fluid reabsorption, peripheral clearance and transporter function at the blood-brain barrier. J Neuroinflammation. 2012;9:150.PubMedCentralCrossRefPubMed Erickson MA, Hartvigson PE, Morofuji Y, Owen JB, Butterfield DA, Banks WA. Lipopolysaccharide impairs amyloid beta efflux from brain: altered vascular sequestration, cerebrospinal fluid reabsorption, peripheral clearance and transporter function at the blood-brain barrier. J Neuroinflammation. 2012;9:150.PubMedCentralCrossRefPubMed
40.
go back to reference Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller MC, Piccio L, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol. 2006;177:3520–4.CrossRefPubMed Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller MC, Piccio L, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol. 2006;177:3520–4.CrossRefPubMed
41.
go back to reference Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM0-2 and DAP12). J Immunol. 2006;177:2051–5.CrossRefPubMed Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM0-2 and DAP12). J Immunol. 2006;177:2051–5.CrossRefPubMed
42.
go back to reference Schmid CD, Melchior B, Masek K, Puntambekar SS, Danielson PE, Lo DD, et al. Differential gene expression in LPS/IFNgamma activated microglia and macrophages: in vitro versus in vivo. Neurochem. 2009;109 Suppl 1:117–25.CrossRef Schmid CD, Melchior B, Masek K, Puntambekar SS, Danielson PE, Lo DD, et al. Differential gene expression in LPS/IFNgamma activated microglia and macrophages: in vitro versus in vivo. Neurochem. 2009;109 Suppl 1:117–25.CrossRef
43.
go back to reference Wispelwey B, Lesse AJ, Hansen EJ, Scheld WM. Haemophilus influenzae lipopolysaccharide-induced blood brain barrier permeability during experimental meningitis in the rat. J Clin Investig. 1988;82:1339–46.PubMedCentralCrossRefPubMed Wispelwey B, Lesse AJ, Hansen EJ, Scheld WM. Haemophilus influenzae lipopolysaccharide-induced blood brain barrier permeability during experimental meningitis in the rat. J Clin Investig. 1988;82:1339–46.PubMedCentralCrossRefPubMed
44.
go back to reference Huber JD, VanGilder RL, Houser KA. Streptozotocin-induced diabetes progressively increases blood-brain barrier permeability in specific brain regions in rats. Am J Physiol. 2006;291:H2660–8. Huber JD, VanGilder RL, Houser KA. Streptozotocin-induced diabetes progressively increases blood-brain barrier permeability in specific brain regions in rats. Am J Physiol. 2006;291:H2660–8.
45.
go back to reference Kim YS, Lee MH, Wisniewski HM. Aluminum induced reversible change in permeability of the blood-brain barrier to [14C]sucrose. Brain Res. 1986;377:286–91.CrossRefPubMed Kim YS, Lee MH, Wisniewski HM. Aluminum induced reversible change in permeability of the blood-brain barrier to [14C]sucrose. Brain Res. 1986;377:286–91.CrossRefPubMed
46.
go back to reference Brown RC, Davis TP. Calcium modulation of adherens tight junction function: a potential mechanism for blood-brain barrier disruption after stroke. Stroke. 2002;33:1706–11.CrossRefPubMed Brown RC, Davis TP. Calcium modulation of adherens tight junction function: a potential mechanism for blood-brain barrier disruption after stroke. Stroke. 2002;33:1706–11.CrossRefPubMed
47.
go back to reference Erickson MA, Banks WA. Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain, Behavior, & Immunity. 2011;25:1637–48.CrossRef Erickson MA, Banks WA. Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain, Behavior, & Immunity. 2011;25:1637–48.CrossRef
48.
go back to reference Farr SA, Poon HF, Dogrukol-Ak D, Drake J, Banks WA, Eyerman E, et al. The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. Journal of Neurochemisrty. 2003;84:1173–83.CrossRef Farr SA, Poon HF, Dogrukol-Ak D, Drake J, Banks WA, Eyerman E, et al. The antioxidants alpha-lipoic acid and N-acetylcysteine reverse memory impairment and brain oxidative stress in aged SAMP8 mice. Journal of Neurochemisrty. 2003;84:1173–83.CrossRef
49.
go back to reference Vane JR, Botting RM. Mechanism of action of anti-inflammatory drugs. Adv Exp Med Biol. 1997;433:133–8. Vane JR, Botting RM. Mechanism of action of anti-inflammatory drugs. Adv Exp Med Biol. 1997;433:133–8.
50.
go back to reference Dohgu S, Banks WA. Lipopolysaccharide-enhanced transcellular transport of HIV-1 across the blood-brain barrier is mediated by the p38 mitogen-activated protein kinase pathway. Exp Neurol. 2008;210:740–9.PubMedCentralCrossRefPubMed Dohgu S, Banks WA. Lipopolysaccharide-enhanced transcellular transport of HIV-1 across the blood-brain barrier is mediated by the p38 mitogen-activated protein kinase pathway. Exp Neurol. 2008;210:740–9.PubMedCentralCrossRefPubMed
51.
go back to reference Engstrom L, Ruud J, Eskilsson A, Larsson A, Mackerlova L, Kugelberg U, et al. Lipopolysaccharide-induced fever depends on prostaglandin E2 production specifically in brain endothelial cells. Endocrinology. 2012;153:4849–61.CrossRefPubMed Engstrom L, Ruud J, Eskilsson A, Larsson A, Mackerlova L, Kugelberg U, et al. Lipopolysaccharide-induced fever depends on prostaglandin E2 production specifically in brain endothelial cells. Endocrinology. 2012;153:4849–61.CrossRefPubMed
52.
go back to reference Inoue W, Matsumura K, Yamagata K, Takemiya T, Shiraki T, Kobayashi S. Brain-specific endothelial induction of prostaglandin E(2) synthesis enzymes and its temporal relation to fever. Neurosci Res. 2002;44:51–61.CrossRefPubMed Inoue W, Matsumura K, Yamagata K, Takemiya T, Shiraki T, Kobayashi S. Brain-specific endothelial induction of prostaglandin E(2) synthesis enzymes and its temporal relation to fever. Neurosci Res. 2002;44:51–61.CrossRefPubMed
53.
go back to reference Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, et al. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007;55:453–62.PubMedCentralCrossRefPubMed Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, et al. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007;55:453–62.PubMedCentralCrossRefPubMed
54.
go back to reference Banks WA, Dohgu S, Nakaoke R, Lynch JL, Fleegal-DeMotta MA, Erickson MA, et al. Nitric oxide isoenzymes regulate LPS-enhanced insulin transport across the blood-brain barrier. Endocrinology. 2008;149:1514–23.PubMedCentralCrossRefPubMed Banks WA, Dohgu S, Nakaoke R, Lynch JL, Fleegal-DeMotta MA, Erickson MA, et al. Nitric oxide isoenzymes regulate LPS-enhanced insulin transport across the blood-brain barrier. Endocrinology. 2008;149:1514–23.PubMedCentralCrossRefPubMed
55.
go back to reference Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV injection. Nat Med. 2006;12:1365–71.CrossRefPubMed Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV injection. Nat Med. 2006;12:1365–71.CrossRefPubMed
56.
go back to reference Ng QY, Lee KW, Byrne C, Ho TF, Lim CL. Plasma endotoxin and immune responses during a 21-km road race under a warm and humid environment. Ann Acad Med Singapore. 2008;37:307–14.PubMed Ng QY, Lee KW, Byrne C, Ho TF, Lim CL. Plasma endotoxin and immune responses during a 21-km road race under a warm and humid environment. Ann Acad Med Singapore. 2008;37:307–14.PubMed
57.
go back to reference Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, et al. Energy intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr. 2008;87:1219–23.PubMed Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, et al. Energy intake is associated with endotoxemia in apparently healthy men. Am J Clin Nutr. 2008;87:1219–23.PubMed
58.
go back to reference Jasarevic E, Howerton CL, Howard CD, Bale TL. Alterations in the vaginal microbiome by maternal stress are associated with metabolic reprogramming of the offspring gut and brain. Endocrinology. 2015;156:3265–76.CrossRefPubMed Jasarevic E, Howerton CL, Howard CD, Bale TL. Alterations in the vaginal microbiome by maternal stress are associated with metabolic reprogramming of the offspring gut and brain. Endocrinology. 2015;156:3265–76.CrossRefPubMed
59.
go back to reference Banks WA. A vagina monologue: mom’s stress, bugs, and baby’s brain. Endocrinology. 2015;156:3066–8.CrossRefPubMed Banks WA. A vagina monologue: mom’s stress, bugs, and baby’s brain. Endocrinology. 2015;156:3066–8.CrossRefPubMed
Metadata
Title
Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit
Authors
William A. Banks
Alicia M. Gray
Michelle A. Erickson
Therese S. Salameh
Mamatha Damodarasamy
Nader Sheibani
James S. Meabon
Emily E. Wing
Yoichi Morofuji
David G. Cook
May J. Reed
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2015
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0434-1

Other articles of this Issue 1/2015

Journal of Neuroinflammation 1/2015 Go to the issue