Aβ1–42-RAGE Interaction Disrupts Tight Junctions of the Blood–Brain Barrier Via Ca2+-Calcineurin Signaling

The blood–brain barrier (BBB), which is formed by adherens and tight junctions (TJs) of endothelial cells, maintains homeostasis of the brain. Disrupted intracellular Ca2+ homeostasis and breakdown of the BBB have been implicated in the pathogenesis of Alzheimer's disease (AD). The receptor for advanced glycation end products (RAGE) is known to interact with amyloid β-peptide (Aβ) and mediate Aβ transport across the BBB, contributing to the deposition of Aβ in the brain. However, molecular mechanisms underlying Aβ-RAGE interaction-induced alterations in the BBB have not been identified. We found that Aβ1–42 induces enhanced permeability, disruption of zonula occludin-1 (ZO-1) expression in the plasma membrane, and increased intracellular calcium and matrix metalloproteinase (MMP) secretion in cultured endothelial cells. Neutralizing antibodies against RAGE and inhibitors of calcineurin and MMPs prevented Aβ1–42-induced changes in ZO-1, suggesting that Aβ-RAGE interactions alter TJ proteins through the Ca2+-calcineurin pathway. Consistent with these in vitro findings, we found disrupted microvessels near Aβ plaque-deposited areas, elevated RAGE expression, and enhanced MMP secretion in microvessels of the brains of 5XFAD mice, an animal model for AD. We have identified a potential molecular pathway underlying Aβ-RAGE interaction-induced breakage of BBB integrity. This pathway might play an important role in the pathogenesis of AD.

[1]  R. Weinberg,et al.  Synaptic Autoregulation by Metalloproteases and γ-Secretase , 2011, The Journal of Neuroscience.

[2]  I. Mook‐Jung,et al.  RAGE regulates BACE1 and Aβ generation via NFAT1 activation in Alzheimer's disease animal model , 2009, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[3]  M. Frosch,et al.  Matrix metalloproteinase inhibition reduces oxidative stress associated with cerebral amyloid angiopathy in vivo in transgenic mice , 2009, Journal of neurochemistry.

[4]  Bo Li,et al.  Amyloid β Interaction with Receptor for Advanced Glycation End Products Up-Regulates Brain Endothelial CCR5 Expression and Promotes T Cells Crossing the Blood-Brain Barrier1 , 2009, The Journal of Immunology.

[5]  I. Romero,et al.  Amyloid-β-induced occludin down-regulation and increased permeability in human brain endothelial cells is mediated by MAPK activation , 2009, Journal of cellular and molecular medicine.

[6]  P. Fraser,et al.  Role of NADPH oxidase in retinal microvascular permeability increase by RAGE activation. , 2009, Investigative ophthalmology & visual science.

[7]  J. Kotarek,et al.  Soluble aggregates of the amyloid‐β protein selectively stimulate permeability in human brain microvascular endothelial monolayers , 2008, Journal of neurochemistry.

[8]  Berislav V. Zlokovic,et al.  New therapeutic targets in the neurovascular pathway in Alzheimer’s disease , 2008, Neurotherapeutics.

[9]  P. Agostinho,et al.  Overactivation of calcineurin induced by amyloid-beta and prion proteins , 2008, Neurochemistry International.

[10]  W. Klein,et al.  Aβ Oligomers Induce Neuronal Oxidative Stress through an N-Methyl-D-aspartate Receptor-dependent Mechanism That Is Blocked by the Alzheimer Drug Memantine* , 2007, Journal of Biological Chemistry.

[11]  Jeffrey F. Thompson,et al.  Matrix Metalloproteinase-Mediated Disruption of Tight Junction Proteins in Cerebral Vessels is Reversed by Synthetic Matrix Metalloproteinase Inhibitor in Focal Ischemia in Rat , 2007, Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.

[12]  J. Pfeilschifter,et al.  Molecular mechanisms of cyclosporin A inhibition of the cytokine-induced matrix metalloproteinase-9 in glomerular mesangial cells. , 2007, Journal of the American Society of Nephrology : JASN.

[13]  Rachel C Brown,et al.  Tight junction protein expression and barrier properties of immortalized mouse brain microvessel endothelial cells , 2007, Brain Research.

[14]  M. Ohno,et al.  Intraneuronal β-Amyloid Aggregates, Neurodegeneration, and Neuron Loss in Transgenic Mice with Five Familial Alzheimer's Disease Mutations: Potential Factors in Amyloid Plaque Formation , 2006, The Journal of Neuroscience.

[15]  S. Skaper,et al.  Amyloid β-peptide1–42 alters tight junction protein distribution and expression in brain microvessel endothelial cells , 2006, Neuroscience Letters.

[16]  K. Tomizawa,et al.  Truncation and Activation of Calcineurin A by Calpain I in Alzheimer Disease Brain* , 2005, Journal of Biological Chemistry.

[17]  T. Davis,et al.  The Blood-Brain Barrier/Neurovascular Unit in Health and Disease , 2005, Pharmacological Reviews.

[18]  S. Hofmann,et al.  Age associated changes of AGE-receptor expression: RAGE upregulation is associated with human heart dysfunction , 2004, Experimental Gerontology.

[19]  D. Holtzman,et al.  Matrix metalloproteinase–9 and spontaneous hemorrhage in an animal model of cerebral amyloid angiopathy , 2003, Annals of neurology.

[20]  Ann Marie Schmidt,et al.  RAGE mediates amyloid-β peptide transport across the blood-brain barrier and accumulation in brain , 2003, Nature Medicine.

[21]  P. Sandercock,et al.  Is Breakdown of the Blood-Brain Barrier Responsible for Lacunar Stroke, Leukoaraiosis, and Dementia? , 2003, Stroke.

[22]  L. K. Baker,et al.  Oligomeric and Fibrillar Species of Amyloid-β Peptides Differentially Affect Neuronal Viability* , 2002, The Journal of Biological Chemistry.

[23]  Rustam Azimov,et al.  The channel hypothesis of Alzheimer’s disease: current status , 2002, Peptides.

[24]  D. Selkoe Alzheimer's disease: genes, proteins, and therapy. , 2001, Physiological reviews.

[25]  Mikio Shoji,et al.  Age-Dependent Changes in Brain, CSF, and Plasma Amyloid β Protein in the Tg2576 Transgenic Mouse Model of Alzheimer's Disease , 2001, The Journal of Neuroscience.

[26]  D. Holtzman,et al.  Clearance of Alzheimer's amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. , 2000, The Journal of clinical investigation.

[27]  Masahiro Kawahara,et al.  Alzheimer's β-Amyloid, Human Islet Amylin, and Prion Protein Fragment Evoke Intracellular Free Calcium Elevations by a Common Mechanism in a Hypothalamic GnRH Neuronal Cell Line* , 2000, The Journal of Biological Chemistry.

[28]  C. Patlak,et al.  In Vitro Evidence That β‐Amyloid Peptide 1–40 Diffuses Across the Blood–Brain Barrier and Affects Its Permeability , 2000, Journal of neuropathology and experimental neurology.

[29]  R. Kalaria The Blood‐Brain Barrier and Cerebrovascular Pathology in Alzheimer's Disease , 1999, Annals of the New York Academy of Sciences.

[30]  E. Matsubara,et al.  Lipoprotein‐free amyloidogenic peptides in plasma are elevated in patients with sporadic Alzheimer's disease and Down's syndrome , 1999, Annals of neurology.

[31]  James M. Anderson,et al.  The Tight Junction Protein ZO-1 Establishes a Link between the Transmembrane Protein Occludin and the Actin Cytoskeleton* , 1998, The Journal of Biological Chemistry.

[32]  A. Newby,et al.  Synergistic upregulation of metalloproteinase‐9 by growth factors and inflammatory cytokines: an absolute requirement for transcription factor NF‐κB , 1998, FEBS letters.

[33]  B. Zlokovic,et al.  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. , 1998, The Journal of clinical investigation.

[34]  S. Snyder,et al.  Immunophilins in the Nervous System , 1998, Neuron.

[35]  R. Mohs,et al.  Cerebral infarcts in patients with autopsy-proven Alzheimer's disease , 1998, Neurology.

[36]  W. Banks,et al.  Interactions of β‐Amyloids with the Blood–Brain Barrier , 1997, Annals of the New York Academy of Sciences.

[37]  A. Heyman,et al.  Consortium to establish a registry for Alzheimer's disease: development, database structure, and selected findings. , 1997, Topics in health information management.

[38]  X. Chen,et al.  RAGE and amyloid-β peptide neurotoxicity in Alzheimer's disease , 1996, Nature.

[39]  R. O. Stuart,et al.  Dependence of Epithelial Intercellular Junction Biogenesis on Thapsigargin-sensitive Intracellular Calcium Stores* , 1996, The Journal of Biological Chemistry.

[40]  Luz Claudio,et al.  Ultrastructural features of the blood-brain barrier in biopsy tissue from Alzheimer's disease patients , 1995, Acta Neuropathologica.

[41]  M. Mattson,et al.  beta-Amyloid peptides destabilize calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity , 1992, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[42]  James M. Anderson,et al.  The tight-junction-specific protein ZO-l is a component of the human and rat blood-brain barriers , 1991, Neuroscience Letters.

[43]  R. Agati,et al.  Leukoaraiosis and dementia. , 1990, Stroke.

[44]  Daniel A. Goodenough,et al.  A FINE STRUCTURAL ANALYSIS OF INTERCELLULAR JUNCTIONS IN THE MOUSE LIVER , 1970, The Journal of cell biology.

[45]  S. Son,et al.  FK506 reduces amyloid plaque burden and induces MMP-9 in AβPP/PS1 double transgenic mice. , 2010, Journal of Alzheimer's disease : JAD.

[46]  S. Skaper,et al.  Amyloid beta-peptide1-42 alters tight junction protein distribution and expression in brain microvessel endothelial cells. , 2006, Neuroscience letters.

[47]  S. Younkin,et al.  Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer's disease. , 2001, The Journal of neuroscience : the official journal of the Society for Neuroscience.

[48]  A. Schmidt,et al.  RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. , 1996, Nature.