Skip to main content
Top
Published in: Metabolic Brain Disease 3/2014

01-09-2014 | Review Article

Brain metabolite clearance: impact on Alzheimer’s disease

Authors: Juan M. Zolezzi, Nibaldo C. Inestrosa

Published in: Metabolic Brain Disease | Issue 3/2014

Login to get access

Abstract

Alzheimer’s Disease (AD) is a complex neurodegenerative disorder often associated with aging and characterized by several critical molecular changes that take place in the brain. Among the molecular hallmarks of AD, increased levels of amyloid β-peptide (Aβ) and the subsequent Aβ-derived damage are the most well-studied factors; however, despite the large amounts of effort and resources devoted to the study of AD and AD pathophysiology, the scientific community still awaits therapeutic alternatives capable of ensuring a better outcome for AD patients. In 2012, Cramer et al. (Science 335:1503-1506 2012) astonished the scientific community by rescuing behavioral and cognitive impairments in AD mouse models via oral administration of bexarotene, a drug used to treat some types of skin cancer. Moreover, these authors demonstrated that bexarotene, a retinoid X receptor (RXR) agonist, exerts major effects on Aβ levels, mainly through increased apolipoprotein E (ApoE) expression. Apart from the valid questions addressed in Cramer’s work, only a few attempts have been made to explain the effects of bexarotene. Most of these explanations have been solely based on the ability of bexarotene to reduce Aβ levels and not on the mechanisms that lead to such a reduction. Although it is well known that an imbalance in the Aβ production/excretion rate is the basis of increased Aβ levels in AD, no further explanations have been proposed to address the potential involvement of the blood-brain barrier (BBB), a critical Aβ-clearance structure, in the bexarotene-mediated effects. Moreover, no attempt has been made to explain how the different effects observed after bexarotene administration are connected to each other. Based on current information and on our own experience with nuclear receptors (NR), we offer new perspectives on the mechanisms of bexarotene action, which should help to improve our knowledge of NRs.
Literature
go back to reference Abramov AY, Canevari L, Duchen MR (2004) β-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci 24:565–575PubMedCrossRef Abramov AY, Canevari L, Duchen MR (2004) β-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci 24:565–575PubMedCrossRef
go back to reference Alzheimer’s Association (2012) Alzheimer’s disease facts and figures. Alzheimers Dement 8:131–168CrossRef Alzheimer’s Association (2012) Alzheimer’s disease facts and figures. Alzheimers Dement 8:131–168CrossRef
go back to reference Barroso E, del Valle J, Porquet D et al (2013) Tau hyperphosphorylation and increased BACE1 and RAGE levels in the cortex of PPARβ/δ-null mice. Biochim Biophys Acta 1832:1241–1248PubMedCrossRef Barroso E, del Valle J, Porquet D et al (2013) Tau hyperphosphorylation and increased BACE1 and RAGE levels in the cortex of PPARβ/δ-null mice. Biochim Biophys Acta 1832:1241–1248PubMedCrossRef
go back to reference Bhel C, Davis JB, Lesley R, Schubert D (1994) Hydrogen peroxide mediates amyloid beta protein toxicity. Cell 77:817–827CrossRef Bhel C, Davis JB, Lesley R, Schubert D (1994) Hydrogen peroxide mediates amyloid beta protein toxicity. Cell 77:817–827CrossRef
go back to reference Carvajal FJ, Inestrosa NC (2011) Interaction of AChE with Aβ aggregates in Alzheimer’s brain: therapeutic relevance of IDN 5706. Front Mol Neurosci 4:19PubMedCentralPubMedCrossRef Carvajal FJ, Inestrosa NC (2011) Interaction of AChE with Aβ aggregates in Alzheimer’s brain: therapeutic relevance of IDN 5706. Front Mol Neurosci 4:19PubMedCentralPubMedCrossRef
go back to reference Caspersen C, Wang N, Yao J et al (2005) Mitochondrial Aβ: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J 19:2040–2041PubMed Caspersen C, Wang N, Yao J et al (2005) Mitochondrial Aβ: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J 19:2040–2041PubMed
go back to reference Cavalluci V, D’Amelio M, Cecconi F (2012) Aβ toxicity in Alzheimer’s disease. Mol Neurobiol 45:366–378CrossRef Cavalluci V, D’Amelio M, Cecconi F (2012) Aβ toxicity in Alzheimer’s disease. Mol Neurobiol 45:366–378CrossRef
go back to reference Chawla A, Boisvert WA, Lee CH et al (2001) A PPAR gamma-LRX-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell 7:161–171PubMedCrossRef Chawla A, Boisvert WA, Lee CH et al (2001) A PPAR gamma-LRX-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell 7:161–171PubMedCrossRef
go back to reference Chen YC, Wu JS, Tsai HD, Huang CY, Chen JJ, Sun GY, Lin TN (2012) Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol Neurobiol. doi:10.1007/s12035-012-8259-8 Chen YC, Wu JS, Tsai HD, Huang CY, Chen JJ, Sun GY, Lin TN (2012) Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol Neurobiol. doi:10.​1007/​s12035-012-8259-8
go back to reference Cho DH, Lee EJ, Kwon KJ, Shin CY, Song KH, Park JH, Han SH (2013) Troglitazone, a thiazolidinedione, decreases tau phosphorylation through the inhibition of cyclin-dependent kinase 5 activity in SH-SY5Y neuroblastoma cells and primary neurons. J Neurochem 126:685–695PubMedCrossRef Cho DH, Lee EJ, Kwon KJ, Shin CY, Song KH, Park JH, Han SH (2013) Troglitazone, a thiazolidinedione, decreases tau phosphorylation through the inhibition of cyclin-dependent kinase 5 activity in SH-SY5Y neuroblastoma cells and primary neurons. J Neurochem 126:685–695PubMedCrossRef
go back to reference Cramer PE, Cirrito JR, Wesson DW et al (2012) ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. Science 335:1503–1506PubMedCentralPubMedCrossRef Cramer PE, Cirrito JR, Wesson DW et al (2012) ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. Science 335:1503–1506PubMedCentralPubMedCrossRef
go back to reference Deane R, Singh I, Sagare AP, Bell RD et al (2012) A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest 122:1377–1392PubMedCentralPubMedCrossRef Deane R, Singh I, Sagare AP, Bell RD et al (2012) A multimodal RAGE-specific inhibitor reduces amyloid β-mediated brain disorder in a mouse model of Alzheimer disease. J Clin Invest 122:1377–1392PubMedCentralPubMedCrossRef
go back to reference Drosatos K, Khan RS, Trent CM, Jiang H, Son NH, Blaner WS, Homma S, Schulze PC, Goldberg IJ (2013) Peroxisome proliferator-activated receptor-γ activation prevents sepsis-related cardiac dysfunction and mortality in mice. Circ Heart Fail 6:550–562PubMedCentralPubMedCrossRef Drosatos K, Khan RS, Trent CM, Jiang H, Son NH, Blaner WS, Homma S, Schulze PC, Goldberg IJ (2013) Peroxisome proliferator-activated receptor-γ activation prevents sepsis-related cardiac dysfunction and mortality in mice. Circ Heart Fail 6:550–562PubMedCentralPubMedCrossRef
go back to reference Dumont M, Stack C, Elipenahli C et al (2012) Bezafibrate administration improves behavioral deficits and tau pathology in P301S mice. Hum Mol Genet 21:5091–5105PubMedCentralPubMedCrossRef Dumont M, Stack C, Elipenahli C et al (2012) Bezafibrate administration improves behavioral deficits and tau pathology in P301S mice. Hum Mol Genet 21:5091–5105PubMedCentralPubMedCrossRef
go back to reference Fantini J, Di Scala C, Yahi N, Troadec JD, Sadelli K, Chahinian H, Garmy N (2014) Bexarotene blocks calcium-permeable ion channels formed by neurotoxic Alzheimer’s β-amyloid peptides. ACS Chem Neurosci. doi:10.1021/cn400183w PubMed Fantini J, Di Scala C, Yahi N, Troadec JD, Sadelli K, Chahinian H, Garmy N (2014) Bexarotene blocks calcium-permeable ion channels formed by neurotoxic Alzheimer’s β-amyloid peptides. ACS Chem Neurosci. doi:10.​1021/​cn400183w PubMed
go back to reference Fuentealba RA, Farias G, Scheu J, Bronfman M, Marzolo MP, Inestrosa NC (2004) Signal transduction during amyloid-β-peptide neurotoxicity: role in Alzheimer disease. Brain Res Brain Res Rev 47:275–289PubMedCrossRef Fuentealba RA, Farias G, Scheu J, Bronfman M, Marzolo MP, Inestrosa NC (2004) Signal transduction during amyloid-β-peptide neurotoxicity: role in Alzheimer disease. Brain Res Brain Res Rev 47:275–289PubMedCrossRef
go back to reference Fuenzalida K, Quintanilla R, Ramos P et al (2007) Peroxisome proliferator-activated receptor γ up-regulates the Bcl-2 anti-apoptotic protein in neurons and induces mitochondrial stabilization and protection against oxidative stress and apoptosis. J Biol Chem 282:37006–37015PubMedCrossRef Fuenzalida K, Quintanilla R, Ramos P et al (2007) Peroxisome proliferator-activated receptor γ up-regulates the Bcl-2 anti-apoptotic protein in neurons and induces mitochondrial stabilization and protection against oxidative stress and apoptosis. J Biol Chem 282:37006–37015PubMedCrossRef
go back to reference Gonzales FJ, Shah YM (2007) PPARα: mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators. Toxicology 246:2–8CrossRef Gonzales FJ, Shah YM (2007) PPARα: mechanism of species differences and hepatocarcinogenesis of peroxisome proliferators. Toxicology 246:2–8CrossRef
go back to reference Gronemeyer H, Gustafsson JA, Laudet V (2004) Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov 3:950–964PubMedCrossRef Gronemeyer H, Gustafsson JA, Laudet V (2004) Principles for modulation of the nuclear receptor superfamily. Nat Rev Drug Discov 3:950–964PubMedCrossRef
go back to reference Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nat Rev Mol Cell Biol 8:101–112PubMedCrossRef Haass C, Selkoe DJ (2007) Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer’s amyloid β-peptide. Nat Rev Mol Cell Biol 8:101–112PubMedCrossRef
go back to reference Haemmerle G, Moustafa T, Woelkart G et al (2011) ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-α and PGC-1α. Nat Med 17:1076–1085PubMedCentralPubMedCrossRef Haemmerle G, Moustafa T, Woelkart G et al (2011) ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-α and PGC-1α. Nat Med 17:1076–1085PubMedCentralPubMedCrossRef
go back to reference Harman FS, Nicol CJ, Marin HE, Ward JM, Gonzales FJ, Peters JM (2004) Peroxisome proliferator-activated receptor-delta attenuates colon carcinogenesis. Nat Med 10:481–483PubMedCrossRef Harman FS, Nicol CJ, Marin HE, Ward JM, Gonzales FJ, Peters JM (2004) Peroxisome proliferator-activated receptor-delta attenuates colon carcinogenesis. Nat Med 10:481–483PubMedCrossRef
go back to reference Hollingshead HE, Killins RL, Borland MG, Girroir EE, Billin AN, Willson TM, Sharma AK, Amin S, Gonzales FJ, Peters JM (2007) Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands do not potentiate growth of human cancer cell lines. Carcinogenesis 28:2641–2649PubMedCrossRef Hollingshead HE, Killins RL, Borland MG, Girroir EE, Billin AN, Willson TM, Sharma AK, Amin S, Gonzales FJ, Peters JM (2007) Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) ligands do not potentiate growth of human cancer cell lines. Carcinogenesis 28:2641–2649PubMedCrossRef
go back to reference Hondares E, Rosell M, Díaz-Delfin J et al (2011) Peroxisome proliferator-activated receptor α (PPARα) induces PPARγ coactivator 1α (PGC-1α) gene expression and contributes to thermogenic activation of brown fat: involvement of PRDM16. J Biol Chem 286:43112–43122PubMedCentralPubMedCrossRef Hondares E, Rosell M, Díaz-Delfin J et al (2011) Peroxisome proliferator-activated receptor α (PPARα) induces PPARγ coactivator 1α (PGC-1α) gene expression and contributes to thermogenic activation of brown fat: involvement of PRDM16. J Biol Chem 286:43112–43122PubMedCentralPubMedCrossRef
go back to reference Hoque MT, Robillard KR, Bendayan R (2012) Regulation of breast cancer resistant protein by peroxisome proliferator-activated receptor α in human brain microvessel endothelial cells. Mol Pharmacol 81:598–609PubMedCrossRef Hoque MT, Robillard KR, Bendayan R (2012) Regulation of breast cancer resistant protein by peroxisome proliferator-activated receptor α in human brain microvessel endothelial cells. Mol Pharmacol 81:598–609PubMedCrossRef
go back to reference Inestrosa NC, Godoy JA, Quintanilla RA, Koenig CS, Bronfman M (2005) Peroxisome proliferator-activated receptor gamma is expressed in hippocampal neurons and its activation prevents β-amyloid neurodegeneration: role of Wnt signaling. Exp Cell Res 304:91–104PubMedCrossRef Inestrosa NC, Godoy JA, Quintanilla RA, Koenig CS, Bronfman M (2005) Peroxisome proliferator-activated receptor gamma is expressed in hippocampal neurons and its activation prevents β-amyloid neurodegeneration: role of Wnt signaling. Exp Cell Res 304:91–104PubMedCrossRef
go back to reference Kalinin S, Richardson JC, Feinstein DL (2009) A PPARdelta agonist reduces amyloid burden and brain inflammation in a transgenic mouse model of Alzheimer’s disease. Curr Alzheimer Res 6:431–437PubMedCrossRef Kalinin S, Richardson JC, Feinstein DL (2009) A PPARdelta agonist reduces amyloid burden and brain inflammation in a transgenic mouse model of Alzheimer’s disease. Curr Alzheimer Res 6:431–437PubMedCrossRef
go back to reference Kang J, Rivest S (2012) Lipid metabolism and neuroinflammation in Alzheimer’s disease: a role for liver X receptors. Endocr Rev 33:715–746PubMedCrossRef Kang J, Rivest S (2012) Lipid metabolism and neuroinflammation in Alzheimer’s disease: a role for liver X receptors. Endocr Rev 33:715–746PubMedCrossRef
go back to reference Kook SY, Hong HS, Moon M, Ha CM, Chang S, Mook-Jung I (2012) Aβ1-42-RAGE interaction disrupts tight junctions of the blood-brain barrier via Ca2+-calcineurin signaling. J Neurosci 32:8845–8854PubMedCrossRef Kook SY, Hong HS, Moon M, Ha CM, Chang S, Mook-Jung I (2012) Aβ1-42-RAGE interaction disrupts tight junctions of the blood-brain barrier via Ca2+-calcineurin signaling. J Neurosci 32:8845–8854PubMedCrossRef
go back to reference LaClair KD, Manaye KF, Lee DL et al (2013) Treatment with bexarotene, a compound that increases apolipoprotein-E, provides no cognitive benefit in mutant APP/PS1 mice. Mol Neurodegener 8:18PubMedCentralPubMedCrossRef LaClair KD, Manaye KF, Lee DL et al (2013) Treatment with bexarotene, a compound that increases apolipoprotein-E, provides no cognitive benefit in mutant APP/PS1 mice. Mol Neurodegener 8:18PubMedCentralPubMedCrossRef
go back to reference LaFerla FM (2012) Preclinical success against Alzheimer’s disease with an old drug. N Engl J Med 367:570–574PubMedCrossRef LaFerla FM (2012) Preclinical success against Alzheimer’s disease with an old drug. N Engl J Med 367:570–574PubMedCrossRef
go back to reference Lee HP, Zhu X, Casadesus G et al (2010) Antioxidant approaches for the treatment of Alzheimer’s disease. Expert Rev Neurother 10:1201–1208PubMedCrossRef Lee HP, Zhu X, Casadesus G et al (2010) Antioxidant approaches for the treatment of Alzheimer’s disease. Expert Rev Neurother 10:1201–1208PubMedCrossRef
go back to reference Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH (2006) Mitochondria are a direct site of Aβ accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 15:1437–1449PubMedCrossRef Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH (2006) Mitochondria are a direct site of Aβ accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 15:1437–1449PubMedCrossRef
go back to reference Mangelsdorf DJ, Borgmeyer U, Heyman RA et al (1992) Characterization of three RXR genes that mediate the action of 9-cis retinoic acid. Genes Dev 6:329–344PubMedCrossRef Mangelsdorf DJ, Borgmeyer U, Heyman RA et al (1992) Characterization of three RXR genes that mediate the action of 9-cis retinoic acid. Genes Dev 6:329–344PubMedCrossRef
go back to reference Manji H, Kato T, Di Prospero NA et al (2012) Impaired mitochondrial function in psychiatric disorders. Nature Rev Neurosci 13:293–307 Manji H, Kato T, Di Prospero NA et al (2012) Impaired mitochondrial function in psychiatric disorders. Nature Rev Neurosci 13:293–307
go back to reference Martín A, Pérez-Girón JV, Hernanz R, Palacios R, Briones AM, Fortuño A, Zalba G, Salaices M, Alonso MJ (2012) Peroxisome proliferator-activated receptor-γ activation reduces cyclooxygenase-2 expression in vascular smooth muscle cells from hypertensive rats by interfering with oxidative stress. J Hypertens 30:315–326PubMedCrossRef Martín A, Pérez-Girón JV, Hernanz R, Palacios R, Briones AM, Fortuño A, Zalba G, Salaices M, Alonso MJ (2012) Peroxisome proliferator-activated receptor-γ activation reduces cyclooxygenase-2 expression in vascular smooth muscle cells from hypertensive rats by interfering with oxidative stress. J Hypertens 30:315–326PubMedCrossRef
go back to reference Meyer-Luehmann M, Spires-Jones TL, Prada C et al (2008) Rapid appearance and local toxicity of amyloid-β plaques in a mouse model of Alzheimer’s disease. Nature 451:720–724PubMedCentralPubMedCrossRef Meyer-Luehmann M, Spires-Jones TL, Prada C et al (2008) Rapid appearance and local toxicity of amyloid-β plaques in a mouse model of Alzheimer’s disease. Nature 451:720–724PubMedCentralPubMedCrossRef
go back to reference Miranda S, Opazo C, Larrondo LF et al (2000) The role of oxidative stress in the toxicity induced by amyloid beta-peptide in Alzheimer’s disease. Prog Neurobiol 62:633–648PubMedCrossRef Miranda S, Opazo C, Larrondo LF et al (2000) The role of oxidative stress in the toxicity induced by amyloid beta-peptide in Alzheimer’s disease. Prog Neurobiol 62:633–648PubMedCrossRef
go back to reference Mulholland DJ, Dedhar S, Coetzee GA, Nelson CC (2005) Interaction of nuclear receptors with the Wnt/beta-catenin/Tcf signaling axis: Wnt you like to know? Endocr Rev 26:898–915PubMedCrossRef Mulholland DJ, Dedhar S, Coetzee GA, Nelson CC (2005) Interaction of nuclear receptors with the Wnt/beta-catenin/Tcf signaling axis: Wnt you like to know? Endocr Rev 26:898–915PubMedCrossRef
go back to reference Muzio G, Maggiora M, Oraldi M, Trombetta A, Canuto RA (2007) PPARalpha and PP2A are involved in the proapoptotic effect of conjugated linoleic acid on human hepatoma cell line SK-HEP-1. Int J Cancer 121:2395–2401PubMedCrossRef Muzio G, Maggiora M, Oraldi M, Trombetta A, Canuto RA (2007) PPARalpha and PP2A are involved in the proapoptotic effect of conjugated linoleic acid on human hepatoma cell line SK-HEP-1. Int J Cancer 121:2395–2401PubMedCrossRef
go back to reference Mysiorek C, Culot M, Dehouck L, Derudas B, Bordet R, Cecchelli R, Fenart L, Berezowski V (2009) Peroxisome-proliferator-activated receptor-alpha activation protects brain capillary endothelial cells from oxygen-glucose deprivation-induced hyperpermeability in the blood-brain barrier. Curr Neurovasc Res 6:181–193PubMedCrossRef Mysiorek C, Culot M, Dehouck L, Derudas B, Bordet R, Cecchelli R, Fenart L, Berezowski V (2009) Peroxisome-proliferator-activated receptor-alpha activation protects brain capillary endothelial cells from oxygen-glucose deprivation-induced hyperpermeability in the blood-brain barrier. Curr Neurovasc Res 6:181–193PubMedCrossRef
go back to reference O’Donnell ME, Lam TI, Tran LQ (2006) Estradiol reduces activity of the blood-brain barrier Na-K-Cl cotransporter and decreases edema formation in permanent middle cerebral artery occlusion. J Cereb Blood Flow Metab 26:1234–1249PubMedCrossRef O’Donnell ME, Lam TI, Tran LQ (2006) Estradiol reduces activity of the blood-brain barrier Na-K-Cl cotransporter and decreases edema formation in permanent middle cerebral artery occlusion. J Cereb Blood Flow Metab 26:1234–1249PubMedCrossRef
go back to reference Paula-Lima AC, Adasme T, SanMartín C et al (2011) Amyloid β-peptide oligomers stimulate RyR-mediated Ca2+ release inducing mitochondrial fragmentation in hippocampal neurons and prevent RyR-mediated dendritic spine remodeling produced by BDNF. Antioxid Redox Signal 14:1209–1223PubMedCrossRef Paula-Lima AC, Adasme T, SanMartín C et al (2011) Amyloid β-peptide oligomers stimulate RyR-mediated Ca2+ release inducing mitochondrial fragmentation in hippocampal neurons and prevent RyR-mediated dendritic spine remodeling produced by BDNF. Antioxid Redox Signal 14:1209–1223PubMedCrossRef
go back to reference Philipson CW, Bassaganya-Riera J, Viladomiu M, Pedragosa M, Guerrant RL, Roche JK, Hontecillas R (2013) The role of peroxisome proliferator-activated receptor γ in immune responses to enteroaggregative Escherichia coli infection. PLoS One 8:e57812PubMedCentralPubMedCrossRef Philipson CW, Bassaganya-Riera J, Viladomiu M, Pedragosa M, Guerrant RL, Roche JK, Hontecillas R (2013) The role of peroxisome proliferator-activated receptor γ in immune responses to enteroaggregative Escherichia coli infection. PLoS One 8:e57812PubMedCentralPubMedCrossRef
go back to reference Reddy OH, Beal MF (2008) Amyloid β, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 14:45–53PubMedCentralPubMedCrossRef Reddy OH, Beal MF (2008) Amyloid β, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 14:45–53PubMedCentralPubMedCrossRef
go back to reference Santos MJ, Quintanilla RA, Toro A, Grandy R, Dinamarca MC, Godoy JA, Inestrosa NC (2005) Peroxisomal proliferation protects from beta-amyloid neurodegeneration. J Biol Chem 280:41057–41068PubMedCrossRef Santos MJ, Quintanilla RA, Toro A, Grandy R, Dinamarca MC, Godoy JA, Inestrosa NC (2005) Peroxisomal proliferation protects from beta-amyloid neurodegeneration. J Biol Chem 280:41057–41068PubMedCrossRef
go back to reference Selkoe DJ (2001) Alzheimer's disease results from the cerebral accumulation and cytotoxicity of amyloid beta-protein. J Alzheimers Dis 3:75–80PubMed Selkoe DJ (2001) Alzheimer's disease results from the cerebral accumulation and cytotoxicity of amyloid beta-protein. J Alzheimers Dis 3:75–80PubMed
go back to reference Selkoe DJ (2011) Resolving controversies on the path to Alzheimer’s therapeuthics. Nat Med 17:1060–1065PubMedCrossRef Selkoe DJ (2011) Resolving controversies on the path to Alzheimer’s therapeuthics. Nat Med 17:1060–1065PubMedCrossRef
go back to reference Sertizing P, Seifert M, Tilgen W, Reichrath J (2007) Present concepts and future outlook: function of peroxisome proliferator-activated receptors (PPARs) for pathogenesis, progression, and therapy of cancer. J Cell Physiol 212:1–12CrossRef Sertizing P, Seifert M, Tilgen W, Reichrath J (2007) Present concepts and future outlook: function of peroxisome proliferator-activated receptors (PPARs) for pathogenesis, progression, and therapy of cancer. J Cell Physiol 212:1–12CrossRef
go back to reference Shaerzadeh F, Motamedi F, Minai-Tehrani D, Khodagholi F (2013) Monitoring of neuronal loss in the hippocampus of Aβ-injected rat: autophagy, mitophagy, and mitochondrial biogenesis stand against apoptosis. Neuromolecular Med. doi:10.1007/s12017-013-8272-8 PubMed Shaerzadeh F, Motamedi F, Minai-Tehrani D, Khodagholi F (2013) Monitoring of neuronal loss in the hippocampus of Aβ-injected rat: autophagy, mitophagy, and mitochondrial biogenesis stand against apoptosis. Neuromolecular Med. doi:10.​1007/​s12017-013-8272-8 PubMed
go back to reference Sheaffer KL, Wada K, Takahashi H, Matsuhashi N, Ohnishi S, Wolfe MM, Turner JR, Nakajima A, Borkan SC, Saubermann LJ (2005) Peroxisome proliferator-activated receptor gamma inhibition prevents adhesion to the extracellular matrix and induces anoikis in hepatocellular carcinoma cells. Cancer Res 65:2251–2259CrossRef Sheaffer KL, Wada K, Takahashi H, Matsuhashi N, Ohnishi S, Wolfe MM, Turner JR, Nakajima A, Borkan SC, Saubermann LJ (2005) Peroxisome proliferator-activated receptor gamma inhibition prevents adhesion to the extracellular matrix and induces anoikis in hepatocellular carcinoma cells. Cancer Res 65:2251–2259CrossRef
go back to reference Silva DF, Selfridge JE, Lu J et al (2013) Bioenergetics flux, mitochondrial mass and mitochondrial morphology dynamics and AD and MCI cybrid cell lines. Hum Mol Genet. doi:10.1093/hmg/ddt247 Silva DF, Selfridge JE, Lu J et al (2013) Bioenergetics flux, mitochondrial mass and mitochondrial morphology dynamics and AD and MCI cybrid cell lines. Hum Mol Genet. doi:10.​1093/​hmg/​ddt247
go back to reference Silva-Alvarez C, Arrázola MS, Godoy JA, Ordenes D, Inestrosa NC (2013) Canonical Wnt signaling protects hippocampal neurons from Aβ oligomers: role of non-canonical Wnt-5ª/Ca(2+) in mitocondrial dynamics. Front Cell Neurosci 7:97PubMedCentralPubMedCrossRef Silva-Alvarez C, Arrázola MS, Godoy JA, Ordenes D, Inestrosa NC (2013) Canonical Wnt signaling protects hippocampal neurons from Aβ oligomers: role of non-canonical Wnt-5ª/Ca(2+) in mitocondrial dynamics. Front Cell Neurosci 7:97PubMedCentralPubMedCrossRef
go back to reference Singh I, Sagare AP, Coma M, Perlmutter D, Gelein R, Bell RD, Deane RJ, Zhong E, Parisi M, Ciszewski J, Kasper RT, Deane R (2013) Low levels of copper disrupt brain amyloid-β homeostasis by altering its production and clearance. PNAS. doi:10.1073/pnas.1302212110 Singh I, Sagare AP, Coma M, Perlmutter D, Gelein R, Bell RD, Deane RJ, Zhong E, Parisi M, Ciszewski J, Kasper RT, Deane R (2013) Low levels of copper disrupt brain amyloid-β homeostasis by altering its production and clearance. PNAS. doi:10.​1073/​pnas.​1302212110
go back to reference Südhof TC (2013) Neurotransmitter release : the last millisecond in the life of a synaptic vesicle. Neuron 80:675–690PubMedCrossRef Südhof TC (2013) Neurotransmitter release : the last millisecond in the life of a synaptic vesicle. Neuron 80:675–690PubMedCrossRef
go back to reference Terry RD, Masliah E, Salmon DP et al (1991) Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 30:572–80PubMedCrossRef Terry RD, Masliah E, Salmon DP et al (1991) Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 30:572–80PubMedCrossRef
go back to reference Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, Ikeuchi T (2012) Hyperphosphorylation of tau induced by naturally secreted amyloid-β at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3β signaling pathway. J Biol Chem 287:35222–35233PubMedCentralPubMedCrossRef Tokutake T, Kasuga K, Yajima R, Sekine Y, Tezuka T, Nishizawa M, Ikeuchi T (2012) Hyperphosphorylation of tau induced by naturally secreted amyloid-β at nanomolar concentrations is modulated by insulin-dependent Akt-GSK3β signaling pathway. J Biol Chem 287:35222–35233PubMedCentralPubMedCrossRef
go back to reference Toledo EM, Inestrosa NC (2010) Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Mol Psychiatry 15:272–285PubMedCrossRef Toledo EM, Inestrosa NC (2010) Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Mol Psychiatry 15:272–285PubMedCrossRef
go back to reference Watson GS, Craft S (2003) The role of insulin resistance in the pathogenesis of Alzheimer’s disease: implications for treatment. CNS Drugs 17:27–45PubMedCrossRef Watson GS, Craft S (2003) The role of insulin resistance in the pathogenesis of Alzheimer’s disease: implications for treatment. CNS Drugs 17:27–45PubMedCrossRef
go back to reference Yin KJ, Deng Z, Hamblin M, Xiang Y, Huang H, Zhang J, Jiang X, Wang Y, Chen YE (2010) Peroxisome proliferator-activated receptor delta regulation of miR-15a in ischemia-induced cerebral vascular endothelial injury. J Neurosci 30:6398–6408PubMedCentralPubMedCrossRef Yin KJ, Deng Z, Hamblin M, Xiang Y, Huang H, Zhang J, Jiang X, Wang Y, Chen YE (2010) Peroxisome proliferator-activated receptor delta regulation of miR-15a in ischemia-induced cerebral vascular endothelial injury. J Neurosci 30:6398–6408PubMedCentralPubMedCrossRef
go back to reference Zlokovic BV (2008) The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57:178–201PubMedCrossRef Zlokovic BV (2008) The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron 57:178–201PubMedCrossRef
go back to reference Zlokovic BV (2011) Neurovascular pathways to neurodegeneration in Alzheimmer’s disease and other disorders. Nat Rev Neurosci 12:723–38PubMedCentralPubMed Zlokovic BV (2011) Neurovascular pathways to neurodegeneration in Alzheimmer’s disease and other disorders. Nat Rev Neurosci 12:723–38PubMedCentralPubMed
Metadata
Title
Brain metabolite clearance: impact on Alzheimer’s disease
Authors
Juan M. Zolezzi
Nibaldo C. Inestrosa
Publication date
01-09-2014
Publisher
Springer US
Published in
Metabolic Brain Disease / Issue 3/2014
Print ISSN: 0885-7490
Electronic ISSN: 1573-7365
DOI
https://doi.org/10.1007/s11011-014-9527-2

Other articles of this Issue 3/2014

Metabolic Brain Disease 3/2014 Go to the issue