Skip to main content
Erschienen in: Metabolic Brain Disease 5/2019

02.07.2019 | Original Article

Amyloid Beta 25–35 induces blood-brain barrier disruption in vitro

verfasst von: Elvis Cuevas, Hector Rosas-Hernandez, Susan M. Burks, Manuel A. Ramirez-Lee, Aida Guzman, Syed Z. Imam, Syed F. Ali, Sumit Sarkar

Erschienen in: Metabolic Brain Disease | Ausgabe 5/2019

Einloggen, um Zugang zu erhalten

Abstract

The amyloid β-peptide (Aβ) is transported across the blood-brain barrier (BBB) by binding with the receptor for advanced glycation end products (RAGE). Previously, we demonstrated that the Aβ fraction 25–35 (Aβ25–35) increases RAGE expression in the rat hippocampus, likely contributing to its neurotoxic effects. However, it is still debated if the interaction of Aβ with RAGE compromises the BBB function in Alzheimer’ disease (AD). Here, we evaluated the effects of Aβ25–35 in an established in vitro model of the BBB. Rat brain microvascular endothelial cells (rBMVECs) were treated with 20 μM active Aβ25–35 or the inactive Aβ35–25 (control), for 24 h. Exposure to Aβ25–35 significantly decreased cell viability, increased cellular necrosis, and increased the production of reactive oxygen species (ROS), which triggered a decrease in the enzyme glutathione peroxidase when compared to the control condition. Aβ25–35 also increased BBB permeability by altering the expression of tight junction proteins (decreasing zonula occludens-1 and increasing occludin). Aβ25–35 induced monolayer disruption and cellular disarrangement of the BBB, with RAGE being highly expressed in the zones of disarrangement. Together, these data suggest that Aβ25–35-induces toxicity by compromising the functionality and integrity of the BBB in vitro.
Literatur
Zurück zum Zitat Alzheimer's A (2016) 2016 Alzheimer's disease facts and figures. Alzheimers Dement 12(4):459–509CrossRef Alzheimer's A (2016) 2016 Alzheimer's disease facts and figures. Alzheimers Dement 12(4):459–509CrossRef
Zurück zum Zitat Bednarczyk J, Lukasiuk K (2011) Tight junctions in neurological diseases. Acta Neurobiol Exp (Wars) 71(4):393–408 Bednarczyk J, Lukasiuk K (2011) Tight junctions in neurological diseases. Acta Neurobiol Exp (Wars) 71(4):393–408
Zurück zum Zitat Bernal-Mondragon C, Rivas-Arancibia S, Kendrick KM, Guevara-Guzman R (2013) Estradiol prevents olfactory dysfunction induced by A-beta 25-35 injection in hippocampus. BMC Neurosci 14:104CrossRefPubMedPubMedCentral Bernal-Mondragon C, Rivas-Arancibia S, Kendrick KM, Guevara-Guzman R (2013) Estradiol prevents olfactory dysfunction induced by A-beta 25-35 injection in hippocampus. BMC Neurosci 14:104CrossRefPubMedPubMedCentral
Zurück zum Zitat Blasig IE, Bellmann C, Cording J, Del Vecchio G, Zwanziger D, Huber O, Haseloff RF (2011) Occludin protein family: oxidative stress and reducing conditions. Antioxid Redox Signal 15(5):1195–1219CrossRefPubMed Blasig IE, Bellmann C, Cording J, Del Vecchio G, Zwanziger D, Huber O, Haseloff RF (2011) Occludin protein family: oxidative stress and reducing conditions. Antioxid Redox Signal 15(5):1195–1219CrossRefPubMed
Zurück zum Zitat Butterfield DA, Kanski J (2002) Methionine residue 35 is critical for the oxidative stress and neurotoxic properties of Alzheimer's amyloid beta-peptide 1-42. Peptides 23(7):1299–1309CrossRefPubMed Butterfield DA, Kanski J (2002) Methionine residue 35 is critical for the oxidative stress and neurotoxic properties of Alzheimer's amyloid beta-peptide 1-42. Peptides 23(7):1299–1309CrossRefPubMed
Zurück zum Zitat Carrano A, Hoozemans JJ, van der Vies SM, Rozemuller AJ, van Horssen J, de Vries HE (2011) Amyloid Beta induces oxidative stress-mediated blood-brain barrier changes in capillary amyloid angiopathy. Antioxid Redox Signal 15(5):1167–1178CrossRefPubMed Carrano A, Hoozemans JJ, van der Vies SM, Rozemuller AJ, van Horssen J, de Vries HE (2011) Amyloid Beta induces oxidative stress-mediated blood-brain barrier changes in capillary amyloid angiopathy. Antioxid Redox Signal 15(5):1167–1178CrossRefPubMed
Zurück zum Zitat Chaney MO, Stine WB, Kokjohn TA, Kuo YM, Esh C, Rahman A, Luehrs DC, Schmidt AM, Stern D, Yan SD, Roher AE (2005) RAGE and amyloid beta interactions: atomic force microscopy and molecular modeling. Biochim Biophys Acta 1741(1–2):199–205CrossRefPubMed Chaney MO, Stine WB, Kokjohn TA, Kuo YM, Esh C, Rahman A, Luehrs DC, Schmidt AM, Stern D, Yan SD, Roher AE (2005) RAGE and amyloid beta interactions: atomic force microscopy and molecular modeling. Biochim Biophys Acta 1741(1–2):199–205CrossRefPubMed
Zurück zum Zitat Cuevas E, Limon D, Perez-Severiano F, Diaz A, Ortega L, Zenteno E, Guevara J (2009) Antioxidant effects of Epicatechin on the hippocampal toxicity caused by amyloid-beta 25-35 in rats. Eur J Pharmacol 616(1–3):122–127CrossRefPubMed Cuevas E, Limon D, Perez-Severiano F, Diaz A, Ortega L, Zenteno E, Guevara J (2009) Antioxidant effects of Epicatechin on the hippocampal toxicity caused by amyloid-beta 25-35 in rats. Eur J Pharmacol 616(1–3):122–127CrossRefPubMed
Zurück zum Zitat Cuevas E, Lantz S, Newport G, Divine B, Wu Q, Paule MG, Tobon-Velasco JC, Ali SF, Santamaria A (2010) On the early toxic effect of quinolinic acid: involvement of RAGE. Neurosci Lett 474(2):74–78CrossRefPubMed Cuevas E, Lantz S, Newport G, Divine B, Wu Q, Paule MG, Tobon-Velasco JC, Ali SF, Santamaria A (2010) On the early toxic effect of quinolinic acid: involvement of RAGE. Neurosci Lett 474(2):74–78CrossRefPubMed
Zurück zum Zitat Cuevas E, Lantz SM, Tobon-Velasco JC, Newport GD, Wu Q, Virmani A, Ali SF, Santamaria A (2011) On the in vivo early toxic properties of A-beta 25-35 peptide in the rat hippocampus: involvement of the receptor-for-advanced glycation-end-products and changes in gene expression. Neurotoxicol Teratol 33(2):288–296CrossRefPubMed Cuevas E, Lantz SM, Tobon-Velasco JC, Newport GD, Wu Q, Virmani A, Ali SF, Santamaria A (2011) On the in vivo early toxic properties of A-beta 25-35 peptide in the rat hippocampus: involvement of the receptor-for-advanced glycation-end-products and changes in gene expression. Neurotoxicol Teratol 33(2):288–296CrossRefPubMed
Zurück zum Zitat Deane RJ (2012) Is RAGE still a therapeutic target for Alzheimer's disease? Future Med Chem 4(7):915–925CrossRefPubMed Deane RJ (2012) Is RAGE still a therapeutic target for Alzheimer's disease? Future Med Chem 4(7):915–925CrossRefPubMed
Zurück zum Zitat Deane R, Zlokovic BV (2007) Role of the blood-brain barrier in the pathogenesis of Alzheimer's disease. Curr Alzheimer Res 4(2):191–197CrossRefPubMed Deane R, Zlokovic BV (2007) Role of the blood-brain barrier in the pathogenesis of Alzheimer's disease. Curr Alzheimer Res 4(2):191–197CrossRefPubMed
Zurück zum Zitat Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B (2003) RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med 9(7):907–913CrossRefPubMed Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B (2003) RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med 9(7):907–913CrossRefPubMed
Zurück zum Zitat Deane R, Wu Z, Zlokovic BV (2004) RAGE (yin) versus LRP (yang) balance regulates alzheimer amyloid beta-peptide clearance through transport across the blood-brain barrier. Stroke 35(11 Suppl 1):2628–2631CrossRefPubMed Deane R, Wu Z, Zlokovic BV (2004) RAGE (yin) versus LRP (yang) balance regulates alzheimer amyloid beta-peptide clearance through transport across the blood-brain barrier. Stroke 35(11 Suppl 1):2628–2631CrossRefPubMed
Zurück zum Zitat Deane R, Singh I, Sagare AP, Bell RD, Ross NT, LaRue B, Love R, Perry S, Paquette N, Deane RJ, Thiyagarajan M, Zarcone T, Fritz G, Friedman AE, Miller BL, Zlokovic BV (2012) A multimodal RAGE-specific inhibitor reduces amyloid beta-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest 122(4):1377–1392CrossRefPubMedPubMedCentral Deane R, Singh I, Sagare AP, Bell RD, Ross NT, LaRue B, Love R, Perry S, Paquette N, Deane RJ, Thiyagarajan M, Zarcone T, Fritz G, Friedman AE, Miller BL, Zlokovic BV (2012) A multimodal RAGE-specific inhibitor reduces amyloid beta-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest 122(4):1377–1392CrossRefPubMedPubMedCentral
Zurück zum Zitat Denizot F, Lang R (1986) Rapid colorimetric assay for cell-growth and survival - modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol Methods 89(2):271–277CrossRefPubMed Denizot F, Lang R (1986) Rapid colorimetric assay for cell-growth and survival - modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J Immunol Methods 89(2):271–277CrossRefPubMed
Zurück zum Zitat Do TM, Dodacki A, Alata W, Calon F, Nicolic S, Scherrmann JM, Farinotti R, Bourasset F (2016) Age-dependent regulation of the blood-brain barrier influx/efflux equilibrium of amyloid-beta peptide in a mouse model of Alzheimer's disease (3xTg-AD). J Alzheimers Dis 49(2):287–300CrossRefPubMed Do TM, Dodacki A, Alata W, Calon F, Nicolic S, Scherrmann JM, Farinotti R, Bourasset F (2016) Age-dependent regulation of the blood-brain barrier influx/efflux equilibrium of amyloid-beta peptide in a mouse model of Alzheimer's disease (3xTg-AD). J Alzheimers Dis 49(2):287–300CrossRefPubMed
Zurück zum Zitat Donahue JE, Flaherty SL, Johanson CE, Duncan JA 3rd, Silverberg GD, Miller MC, Tavares R, Yang W, Wu Q, Sabo E, Hovanesian V, Stopa EG (2006) RAGE, LRP-1, and amyloid-beta protein in Alzheimer's disease. Acta Neuropathol 112(4):405–415CrossRefPubMed Donahue JE, Flaherty SL, Johanson CE, Duncan JA 3rd, Silverberg GD, Miller MC, Tavares R, Yang W, Wu Q, Sabo E, Hovanesian V, Stopa EG (2006) RAGE, LRP-1, and amyloid-beta protein in Alzheimer's disease. Acta Neuropathol 112(4):405–415CrossRefPubMed
Zurück zum Zitat Fanning AS, Jameson BJ, Jesaitis LA, Anderson JM (1998) The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton. J Biol Chem 273(45):29745–29753CrossRefPubMed Fanning AS, Jameson BJ, Jesaitis LA, Anderson JM (1998) The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton. J Biol Chem 273(45):29745–29753CrossRefPubMed
Zurück zum Zitat Frozza RL, Horn AP, Hoppe JB, Simao F, Gerhardt D, Comiran RA, Salbego CG (2009) A comparative study of beta-amyloid peptides Abeta1-42 and Abeta25-35 toxicity in organotypic hippocampal slice cultures. Neurochem Res 34(2):295–303CrossRefPubMed Frozza RL, Horn AP, Hoppe JB, Simao F, Gerhardt D, Comiran RA, Salbego CG (2009) A comparative study of beta-amyloid peptides Abeta1-42 and Abeta25-35 toxicity in organotypic hippocampal slice cultures. Neurochem Res 34(2):295–303CrossRefPubMed
Zurück zum Zitat Gheorghiu M, Enciu AM, Popescu BO, Gheorghiu E (2014) Functional and molecular characterization of the effect of amyloid-beta42 on an in vitro epithelial barrier model. J Alzheimers Dis 38(4):787–798CrossRefPubMed Gheorghiu M, Enciu AM, Popescu BO, Gheorghiu E (2014) Functional and molecular characterization of the effect of amyloid-beta42 on an in vitro epithelial barrier model. J Alzheimers Dis 38(4):787–798CrossRefPubMed
Zurück zum Zitat Gospodarska E, Kupniewska-Kozak A, Goch G, Dadlez M (2011) Binding studies of truncated variants of the Abeta peptide to the V-domain of the RAGE receptor reveal Abeta residues responsible for binding. Biochim Biophys Acta 1814(5):592–609CrossRefPubMed Gospodarska E, Kupniewska-Kozak A, Goch G, Dadlez M (2011) Binding studies of truncated variants of the Abeta peptide to the V-domain of the RAGE receptor reveal Abeta residues responsible for binding. Biochim Biophys Acta 1814(5):592–609CrossRefPubMed
Zurück zum Zitat Guimaraes CC, Oliveira DD, Valdevite M, Saltoratto AL, Pereira SI, Franca Sde C, Pereira AM, Pereira PS (2015) The glycosylated flavonoids vitexin, isovitexin, and quercetrin isolated from Serjania erecta Radlk (Sapindaceae) leaves protect PC12 cells against amyloid-beta25-35 peptide-induced toxicity. Food Chem Toxicol 86:88–94CrossRefPubMed Guimaraes CC, Oliveira DD, Valdevite M, Saltoratto AL, Pereira SI, Franca Sde C, Pereira AM, Pereira PS (2015) The glycosylated flavonoids vitexin, isovitexin, and quercetrin isolated from Serjania erecta Radlk (Sapindaceae) leaves protect PC12 cells against amyloid-beta25-35 peptide-induced toxicity. Food Chem Toxicol 86:88–94CrossRefPubMed
Zurück zum Zitat Hardy J, Allsop D (1991) Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends Pharmacol Sci 12(10):383–388CrossRefPubMed Hardy J, Allsop D (1991) Amyloid deposition as the central event in the aetiology of Alzheimer's disease. Trends Pharmacol Sci 12(10):383–388CrossRefPubMed
Zurück zum Zitat Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297(5580):353–356CrossRefPubMed Hardy J, Selkoe DJ (2002) The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297(5580):353–356CrossRefPubMed
Zurück zum Zitat Kook SY, Hong HS, Moon M, Ha CM, Chang S, Mook-Jung I (2012) Abeta(1)(−)(4)(2)-RAGE interaction disrupts tight junctions of the blood-brain barrier via ca(2)(+)-calcineurin signaling. J Neurosci 32(26):8845–8854CrossRefPubMedPubMedCentral Kook SY, Hong HS, Moon M, Ha CM, Chang S, Mook-Jung I (2012) Abeta(1)(−)(4)(2)-RAGE interaction disrupts tight junctions of the blood-brain barrier via ca(2)(+)-calcineurin signaling. J Neurosci 32(26):8845–8854CrossRefPubMedPubMedCentral
Zurück zum Zitat Kubo T, Nishimura S, Kumagae Y, Kaneko I (2002) In vivo conversion of racemized beta-amyloid ([D-Ser 26]A beta 1-40) to truncated and toxic fragments ([D-Ser 26]A beta 25-35/40) and fragment presence in the brains of Alzheimer's patients. J Neurosci Res 70:474–483 Kubo T, Nishimura S, Kumagae Y, Kaneko I (2002) In vivo conversion of racemized beta-amyloid ([D-Ser 26]A beta 1-40) to truncated and toxic fragments ([D-Ser 26]A beta 25-35/40) and fragment presence in the brains of Alzheimer's patients. J Neurosci Res 70:474–483
Zurück zum Zitat Liu R, Gao M, Qiang GF, Zhang TT, Lan X, Ying J, Du GH (2009) The anti-amnesic effects of luteolin against amyloid beta(25-35) peptide-induced toxicity in mice involve the protection of neurovascular unit. Neuroscience 162(4):1232–1243CrossRefPubMed Liu R, Gao M, Qiang GF, Zhang TT, Lan X, Ying J, Du GH (2009) The anti-amnesic effects of luteolin against amyloid beta(25-35) peptide-induced toxicity in mice involve the protection of neurovascular unit. Neuroscience 162(4):1232–1243CrossRefPubMed
Zurück zum Zitat Liu T, Liu WH, Zhao JS, Meng FZ, Wang H (2017a) Lutein protects against beta-amyloid peptide-induced oxidative stress in cerebrovascular endothelial cells through modulation of Nrf-2 and NF-kappab. Cell Biol Toxicol 33(1):57–67CrossRefPubMed Liu T, Liu WH, Zhao JS, Meng FZ, Wang H (2017a) Lutein protects against beta-amyloid peptide-induced oxidative stress in cerebrovascular endothelial cells through modulation of Nrf-2 and NF-kappab. Cell Biol Toxicol 33(1):57–67CrossRefPubMed
Zurück zum Zitat Liu XY, Zhang LJ, Chen Z, Liu LB (2017b) The PTEN inhibitor bpV(pic) promotes neuroprotection against amyloid beta-peptide (25-35)-induced oxidative stress and neurotoxicity. Neurol Res 39(8):758–765CrossRefPubMed Liu XY, Zhang LJ, Chen Z, Liu LB (2017b) The PTEN inhibitor bpV(pic) promotes neuroprotection against amyloid beta-peptide (25-35)-induced oxidative stress and neurotoxicity. Neurol Res 39(8):758–765CrossRefPubMed
Zurück zum Zitat Lochhead JJ, McCaffrey G, Quigley CE, Finch J, DeMarco KM, Nametz N, Davis TP (2010) Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation. J Cereb Blood Flow Metab 30(9):1625–1636CrossRefPubMedPubMedCentral Lochhead JJ, McCaffrey G, Quigley CE, Finch J, DeMarco KM, Nametz N, Davis TP (2010) Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation. J Cereb Blood Flow Metab 30(9):1625–1636CrossRefPubMedPubMedCentral
Zurück zum Zitat Mackic JB, Stins M, McComb JG, Calero M, Ghiso J, Kim KS, Yan SD, Stern D, Schmidt AM, Frangione B, Zlokovic BV (1998) Human blood-brain barrier receptors for Alzheimer's amyloid-beta 1- 40. Asymmetrical binding, endocytosis, and transcytosis at the apical side of brain microvascular endothelial cell monolayer. J Clin Invest 102(4):734–743CrossRefPubMedPubMedCentral Mackic JB, Stins M, McComb JG, Calero M, Ghiso J, Kim KS, Yan SD, Stern D, Schmidt AM, Frangione B, Zlokovic BV (1998) Human blood-brain barrier receptors for Alzheimer's amyloid-beta 1- 40. Asymmetrical binding, endocytosis, and transcytosis at the apical side of brain microvascular endothelial cell monolayer. J Clin Invest 102(4):734–743CrossRefPubMedPubMedCentral
Zurück zum Zitat Mark KS, Davis TP (2002) Cerebral microvascular changes in permeability and tight junctions induced by hypoxia-reoxygenation. Am J Physiol Heart Circ Physiol 282(4):H1485–H1494CrossRefPubMed Mark KS, Davis TP (2002) Cerebral microvascular changes in permeability and tight junctions induced by hypoxia-reoxygenation. Am J Physiol Heart Circ Physiol 282(4):H1485–H1494CrossRefPubMed
Zurück zum Zitat Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ (2010) Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330(6012):1774CrossRefPubMedPubMedCentral Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, Yarasheski KE, Bateman RJ (2010) Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330(6012):1774CrossRefPubMedPubMedCentral
Zurück zum Zitat Millucci L, Ghezzi L, Bernardini G, Santucci A (2010) Conformations and biological activities of amyloid beta peptide 25-35. Curr Protein Pept Sci 11:54–67 Millucci L, Ghezzi L, Bernardini G, Santucci A (2010) Conformations and biological activities of amyloid beta peptide 25-35. Curr Protein Pept Sci 11:54–67
Zurück zum Zitat Paik S, Somvanshi RK, Kumar U (2018) Somatostatin maintains permeability and integrity of blood-brain barrier in beta-amyloid induced toxicity. Mol Neurobiol Paik S, Somvanshi RK, Kumar U (2018) Somatostatin maintains permeability and integrity of blood-brain barrier in beta-amyloid induced toxicity. Mol Neurobiol
Zurück zum Zitat Park S, Kim DS, Kang S, Moon NR (2013) Beta-amyloid-induced cognitive dysfunction impairs glucose homeostasis by increasing insulin resistance and decreasing beta-cell mass in non-diabetic and diabetic rats. Metabolism 62(12):1749–1760CrossRefPubMed Park S, Kim DS, Kang S, Moon NR (2013) Beta-amyloid-induced cognitive dysfunction impairs glucose homeostasis by increasing insulin resistance and decreasing beta-cell mass in non-diabetic and diabetic rats. Metabolism 62(12):1749–1760CrossRefPubMed
Zurück zum Zitat Perez-Severiano F, Salvatierra-Sanchez R, Rodriguez-Perez M, Cuevas-Martinez EY, Guevara J, Limon D, Maldonado PD, Medina-Campos ON, Pedraza-Chaverri J, Santamaria A (2004) S-allylcysteine prevents amyloid-beta peptide-induced oxidative stress in rat hippocampus and ameliorates learning deficits. Eur J Pharmacol 489(3):197–202CrossRefPubMed Perez-Severiano F, Salvatierra-Sanchez R, Rodriguez-Perez M, Cuevas-Martinez EY, Guevara J, Limon D, Maldonado PD, Medina-Campos ON, Pedraza-Chaverri J, Santamaria A (2004) S-allylcysteine prevents amyloid-beta peptide-induced oxidative stress in rat hippocampus and ameliorates learning deficits. Eur J Pharmacol 489(3):197–202CrossRefPubMed
Zurück zum Zitat Pike CJ, Burdick D, Walencewicz AJ, Glabe CG, Cotman CW (1993) Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J Neurosci 13(4):1676–1687CrossRefPubMedPubMedCentral Pike CJ, Burdick D, Walencewicz AJ, Glabe CG, Cotman CW (1993) Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J Neurosci 13(4):1676–1687CrossRefPubMedPubMedCentral
Zurück zum Zitat Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, Cribbs DH, Glabe CG, Cotman CW (1995) Structure-activity analyses of beta-amyloid peptides: contributions of the beta 25-35 region to aggregation and neurotoxicity. J Neurochem 64(1):253–265CrossRefPubMed Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, Cribbs DH, Glabe CG, Cotman CW (1995) Structure-activity analyses of beta-amyloid peptides: contributions of the beta 25-35 region to aggregation and neurotoxicity. J Neurochem 64(1):253–265CrossRefPubMed
Zurück zum Zitat Provias J, Jeynes B (2014) The role of the blood-brain barrier in the pathogenesis of senile plaques in Alzheimer's disease. Int J Alzheimers Dis 2014:191863PubMedPubMedCentral Provias J, Jeynes B (2014) The role of the blood-brain barrier in the pathogenesis of senile plaques in Alzheimer's disease. Int J Alzheimers Dis 2014:191863PubMedPubMedCentral
Zurück zum Zitat Qosa H, LeVine H 3rd, Keller JN, Kaddoumi A (2014) Mixed oligomers and monomeric amyloid-beta disrupts endothelial cells integrity and reduces monomeric amyloid-beta transport across hCMEC/D3 cell line as an in vitro blood-brain barrier model. Biochim Biophys Acta 1842(9):1806–1815CrossRefPubMedPubMedCentral Qosa H, LeVine H 3rd, Keller JN, Kaddoumi A (2014) Mixed oligomers and monomeric amyloid-beta disrupts endothelial cells integrity and reduces monomeric amyloid-beta transport across hCMEC/D3 cell line as an in vitro blood-brain barrier model. Biochim Biophys Acta 1842(9):1806–1815CrossRefPubMedPubMedCentral
Zurück zum Zitat Rosas-Hernandez H, Cuevas E, Lantz SM, Hamilton WR, Ramirez-Lee MA, Ali SF, Gonzalez C (2013a) Prolactin and blood-brain barrier permeability. Curr Neurovasc Res 10(4):278–286CrossRefPubMed Rosas-Hernandez H, Cuevas E, Lantz SM, Hamilton WR, Ramirez-Lee MA, Ali SF, Gonzalez C (2013a) Prolactin and blood-brain barrier permeability. Curr Neurovasc Res 10(4):278–286CrossRefPubMed
Zurück zum Zitat Rosas-Hernandez H, Cuevas E, Lantz-McPeak SM, Ali SF, Gonzalez C (2013b) Prolactin protects against the methamphetamine-induced cerebral vascular toxicity. Curr Neurovasc Res 10(4):346–355CrossRefPubMed Rosas-Hernandez H, Cuevas E, Lantz-McPeak SM, Ali SF, Gonzalez C (2013b) Prolactin protects against the methamphetamine-induced cerebral vascular toxicity. Curr Neurovasc Res 10(4):346–355CrossRefPubMed
Zurück zum Zitat Rosas-Hernandez H, Cuevas E, Escudero-Lourdes C, Lantz SM, Sturdivant NM, Imam SZ, Sarkar S, Slikker W Jr, Paule MG, Balachandran K, Ali SF (2018a) Characterization of uniaxial high-speed stretch as an in vitro model of mild traumatic brain injury on the blood-brain barrier. Neurosci Lett 672:123–129CrossRefPubMed Rosas-Hernandez H, Cuevas E, Escudero-Lourdes C, Lantz SM, Sturdivant NM, Imam SZ, Sarkar S, Slikker W Jr, Paule MG, Balachandran K, Ali SF (2018a) Characterization of uniaxial high-speed stretch as an in vitro model of mild traumatic brain injury on the blood-brain barrier. Neurosci Lett 672:123–129CrossRefPubMed
Zurück zum Zitat Rosas-Hernandez H, Cuevas E, Lantz SM, Paule MG, Ali SF (2018b) Isolation and culture of brain microvascular endothelial cells for in vitro blood-brain barrier studies. Methods Mol Biol 1727:315–331CrossRefPubMed Rosas-Hernandez H, Cuevas E, Lantz SM, Paule MG, Ali SF (2018b) Isolation and culture of brain microvascular endothelial cells for in vitro blood-brain barrier studies. Methods Mol Biol 1727:315–331CrossRefPubMed
Zurück zum Zitat Self RL, Smith KJ, Mulholland PJ, Prendergast MA (2005) Ethanol exposure and withdrawal sensitizes the rat hippocampal CA1 pyramidal cell region to beta-amyloid (25-35)-induced cytotoxicity: NMDA receptor involvement. Alcohol Clin Exp Res 29(11):2063–2069CrossRefPubMed Self RL, Smith KJ, Mulholland PJ, Prendergast MA (2005) Ethanol exposure and withdrawal sensitizes the rat hippocampal CA1 pyramidal cell region to beta-amyloid (25-35)-induced cytotoxicity: NMDA receptor involvement. Alcohol Clin Exp Res 29(11):2063–2069CrossRefPubMed
Zurück zum Zitat Steinerman JR, Irizarry M, Scarmeas N, Raju S, Brandt J, Albert M, Blacker D, Hyman B, Stern Y (2008) Distinct pools of beta-amyloid in Alzheimer disease-affected brain: a clinicopathologic study. Arch Neurol 65(7):906–912CrossRefPubMedPubMedCentral Steinerman JR, Irizarry M, Scarmeas N, Raju S, Brandt J, Albert M, Blacker D, Hyman B, Stern Y (2008) Distinct pools of beta-amyloid in Alzheimer disease-affected brain: a clinicopathologic study. Arch Neurol 65(7):906–912CrossRefPubMedPubMedCentral
Zurück zum Zitat Stepanichev MY, Moiseeva YV, Lazareva NA, Onufriev MV, Gulyaeva NV (2010) Changes in cell proliferation in the subventricular zone of the brain in adult rats given beta-amyloid peptide (25-35). Neurosci Behav Physiol 40(2):123–126CrossRefPubMed Stepanichev MY, Moiseeva YV, Lazareva NA, Onufriev MV, Gulyaeva NV (2010) Changes in cell proliferation in the subventricular zone of the brain in adult rats given beta-amyloid peptide (25-35). Neurosci Behav Physiol 40(2):123–126CrossRefPubMed
Zurück zum Zitat Strazielle N, Ghersi-Egea JF, Ghiso J, Dehouck MP, Frangione B, Patlak C, Fenstermacher J, Gorevic P (2000) In vitro evidence that beta-amyloid peptide 1-40 diffuses across the blood-brain barrier and affects its permeability. J Neuropathol Exp Neurol 59(1):29–38CrossRefPubMed Strazielle N, Ghersi-Egea JF, Ghiso J, Dehouck MP, Frangione B, Patlak C, Fenstermacher J, Gorevic P (2000) In vitro evidence that beta-amyloid peptide 1-40 diffuses across the blood-brain barrier and affects its permeability. J Neuropathol Exp Neurol 59(1):29–38CrossRefPubMed
Zurück zum Zitat Thal DR, Rub U, Orantes M, Braak H (2002) Phases of a beta-deposition in the human brain and its relevance for the development of AD. Neurology 58(12):1791–1800CrossRefPubMed Thal DR, Rub U, Orantes M, Braak H (2002) Phases of a beta-deposition in the human brain and its relevance for the development of AD. Neurology 58(12):1791–1800CrossRefPubMed
Zurück zum Zitat Uttara B, Singh AV, Zamboni P, Mahajan RT (2009) Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol 7(1):65–74CrossRefPubMedPubMedCentral Uttara B, Singh AV, Zamboni P, Mahajan RT (2009) Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options. Curr Neuropharmacol 7(1):65–74CrossRefPubMedPubMedCentral
Zurück zum Zitat Wan WB, Cao L, Liu LM, Kalionis B, Chen C, Tai XT, Li YM, Xia SJ (2014a) EGb761 provides a protective effect against Abeta1-42 oligomer-induced cell damage and blood-brain barrier disruption in an in vitro bEnd.3 endothelial model. PLoS One 9(11):e113126CrossRefPubMedPubMedCentral Wan WB, Cao L, Liu LM, Kalionis B, Chen C, Tai XT, Li YM, Xia SJ (2014a) EGb761 provides a protective effect against Abeta1-42 oligomer-induced cell damage and blood-brain barrier disruption in an in vitro bEnd.3 endothelial model. PLoS One 9(11):e113126CrossRefPubMedPubMedCentral
Zurück zum Zitat Wan W, Chen H, Li Y (2014b) The potential mechanisms of Abeta-receptor for advanced glycation end-products interaction disrupting tight junctions of the blood-brain barrier in Alzheimer's disease. Int J Neurosci 124(2):75–81CrossRefPubMed Wan W, Chen H, Li Y (2014b) The potential mechanisms of Abeta-receptor for advanced glycation end-products interaction disrupting tight junctions of the blood-brain barrier in Alzheimer's disease. Int J Neurosci 124(2):75–81CrossRefPubMed
Zurück zum Zitat Wan W, Cao L, Liu L, Zhang C, Kalionis B, Tai X, Li Y, Xia S (2015) Abeta(1-42) oligomer-induced leakage in an in vitro blood-brain barrier model is associated with up-regulation of RAGE and metalloproteinases, and down-regulation of tight junction scaffold proteins. J Neurochem 134(2):382–393CrossRefPubMed Wan W, Cao L, Liu L, Zhang C, Kalionis B, Tai X, Li Y, Xia S (2015) Abeta(1-42) oligomer-induced leakage in an in vitro blood-brain barrier model is associated with up-regulation of RAGE and metalloproteinases, and down-regulation of tight junction scaffold proteins. J Neurochem 134(2):382–393CrossRefPubMed
Zurück zum Zitat Wang H, Joseph JA (1999) Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med 27(5–6):612–616CrossRefPubMed Wang H, Joseph JA (1999) Quantifying cellular oxidative stress by dichlorofluorescein assay using microplate reader. Free Radic Biol Med 27(5–6):612–616CrossRefPubMed
Zurück zum Zitat Wang H, Chen F, Du YF, Long Y, Reed MN, Hu M, Suppiramaniam V, Hong H, Tang SS (2018) Targeted inhibition of RAGE reduces amyloid-beta influx across the blood-brain barrier and improves cognitive deficits in db/db mice. Neuropharmacology 131:143–153CrossRefPubMed Wang H, Chen F, Du YF, Long Y, Reed MN, Hu M, Suppiramaniam V, Hong H, Tang SS (2018) Targeted inhibition of RAGE reduces amyloid-beta influx across the blood-brain barrier and improves cognitive deficits in db/db mice. Neuropharmacology 131:143–153CrossRefPubMed
Zurück zum Zitat Watson PM, Anderson JM, Vanltallie CM, Doctrow SR (1991) The tight-junction-specific protein ZO-1 is a component of the human and rat blood-brain barriers. Neurosci Lett 129(1):6–10CrossRefPubMed Watson PM, Anderson JM, Vanltallie CM, Doctrow SR (1991) The tight-junction-specific protein ZO-1 is a component of the human and rat blood-brain barriers. Neurosci Lett 129(1):6–10CrossRefPubMed
Zurück zum Zitat Weller RO, Subash M, Preston SD, Mazanti I, Carare RO (2008) Perivascular drainage of amyloid-beta peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer's disease. Brain Pathol 18(2):253–266CrossRefPubMed Weller RO, Subash M, Preston SD, Mazanti I, Carare RO (2008) Perivascular drainage of amyloid-beta peptides from the brain and its failure in cerebral amyloid angiopathy and Alzheimer's disease. Brain Pathol 18(2):253–266CrossRefPubMed
Zurück zum Zitat Xie J, Reverdatto S, Frolov A, Hoffmann R, Burz DS, Shekhtman A (2008) Structural basis for pattern recognition by the receptor for advanced glycation end products (RAGE). J Biol Chem 283(40):27255–27269CrossRefPubMed Xie J, Reverdatto S, Frolov A, Hoffmann R, Burz DS, Shekhtman A (2008) Structural basis for pattern recognition by the receptor for advanced glycation end products (RAGE). J Biol Chem 283(40):27255–27269CrossRefPubMed
Zurück zum Zitat Xu P, Wang H, Li Z, Yang Z (2016) Triptolide attenuated injury via inhibiting oxidative stress in amyloid-Beta25-35-treated differentiated PC12 cells. Life Sci 145:19–26CrossRefPubMed Xu P, Wang H, Li Z, Yang Z (2016) Triptolide attenuated injury via inhibiting oxidative stress in amyloid-Beta25-35-treated differentiated PC12 cells. Life Sci 145:19–26CrossRefPubMed
Zurück zum Zitat Yan SD, Chen X, Fu J, Chen M, Godman G, Stern D, Schmidt AM (1996a) RAGE: a receptor upregulated in Alzheimer's disease on neurons, microglia, and cerebrovascular endothelium that binds amyloid-beta peptide and mediates induction of oxidant stress. Neurology 46(2):23005–23005 Yan SD, Chen X, Fu J, Chen M, Godman G, Stern D, Schmidt AM (1996a) RAGE: a receptor upregulated in Alzheimer's disease on neurons, microglia, and cerebrovascular endothelium that binds amyloid-beta peptide and mediates induction of oxidant stress. Neurology 46(2):23005–23005
Zurück zum Zitat Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery T, Zhao L, Nagashima M, Morser J, Migheli A, Nawroth P, Stern D, Schmidt AM (1996b) RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. Nature 382(6593):685–691CrossRefPubMed Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery T, Zhao L, Nagashima M, Morser J, Migheli A, Nawroth P, Stern D, Schmidt AM (1996b) RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. Nature 382(6593):685–691CrossRefPubMed
Zurück zum Zitat Yankner BA, Duffy LK, Kirschner DA (1990) Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides. Science 250(4978):279–282CrossRefPubMed Yankner BA, Duffy LK, Kirschner DA (1990) Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides. Science 250(4978):279–282CrossRefPubMed
Zurück zum Zitat Zhang JX, Xing JG, Wang LL, Jiang HL, Guo SL, Liu R (2017) Luteolin inhibits fibrillary beta-Amyloid1-40-induced inflammation in a human blood-brain barrier model by suppressing the p38 MAPK-mediated NF-kappaB signaling pathways. Molecules 22(3) Zhang JX, Xing JG, Wang LL, Jiang HL, Guo SL, Liu R (2017) Luteolin inhibits fibrillary beta-Amyloid1-40-induced inflammation in a human blood-brain barrier model by suppressing the p38 MAPK-mediated NF-kappaB signaling pathways. Molecules 22(3)
Zurück zum Zitat Zheng X, Xie Z, Zhu Z, Liu Z, Wang Y, Wei L, Yang H, Yang H, Liu Y, Bi J (2014) Methyllycaconitine alleviates amyloid-beta peptides-induced cytotoxicity in SH-SY5Y cells. PLoS One 9(10):e111536CrossRefPubMedPubMedCentral Zheng X, Xie Z, Zhu Z, Liu Z, Wang Y, Wei L, Yang H, Yang H, Liu Y, Bi J (2014) Methyllycaconitine alleviates amyloid-beta peptides-induced cytotoxicity in SH-SY5Y cells. PLoS One 9(10):e111536CrossRefPubMedPubMedCentral
Metadaten
Titel
Amyloid Beta 25–35 induces blood-brain barrier disruption in vitro
verfasst von
Elvis Cuevas
Hector Rosas-Hernandez
Susan M. Burks
Manuel A. Ramirez-Lee
Aida Guzman
Syed Z. Imam
Syed F. Ali
Sumit Sarkar
Publikationsdatum
02.07.2019
Verlag
Springer US
Erschienen in
Metabolic Brain Disease / Ausgabe 5/2019
Print ISSN: 0885-7490
Elektronische ISSN: 1573-7365
DOI
https://doi.org/10.1007/s11011-019-00447-8

Weitere Artikel der Ausgabe 5/2019

Metabolic Brain Disease 5/2019 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Neurologie

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