Oxidative stress and the amyloid beta peptide in Alzheimer’s disease

Redox Biology - Tập 14 - Trang 450-464 - 2018
Clémence Cheignon1,2,3, Mireia Tomas1,2,3, Dominique Bonnefont‐Rousselot4,5,6, Peter Faller7, Christelle Hureau1,2,3, Fabrice Collin1,2,3
1LCC - Laboratoire de chimie de coordination (205 Route de Narbonne 31077 TOULOUSE CEDEX 4 - France)
2Toulouse INP - Institut National Polytechnique (Toulouse) (France)
3UT3 - Université Toulouse III - Paul Sabatier (118 route de Narbonne - 31062 Toulouse - France)
4CHU Pitié-Salpêtrière [AP-HP] (47-83 Boulevard de l'Hôpital, 75013 Paris - France)
5UPD5 - Université Paris Descartes - Paris 5 (12, rue de l'École de Médecine - 75270 Paris cedex 06 - France)
6UTCBS - UM 4 (UMR 8258 / U1022) - Unité de Technologies Chimiques et Biologiques pour la Santé (Faculté des Sciences Pharmaceutiques et Biologiques 4, av de l’Observatoire 75006 PARIS - France)
7IC - Institut de Chimie de Strasbourg (1 rue Blaise Pascal BP 296R8 67008 STRASBOURG CEDEX - France)

Tóm tắt

Từ khóa


Tài liệu tham khảo

Sousa, 2013, Early bioenergetic evolution, Philos. Trans. R. Soc. B: Biol. Sci., 368, 10.1098/rstb.2013.0088

Gorrini, 2013, Modulation of oxidative stress as an anticancer strategy, Nat. Rev. Drug Discov., 12, 931, 10.1038/nrd4002

Halliwell, 1989

Chassaing, 2012, Copper and heme-mediated abeta toxicity: redox chemistry, abeta oxidations and anti-ROS compounds, Curr. Top. Med. Chem., 12, 2573, 10.2174/1568026611212220011

Halliwell, 2006, Oxidative stress and neurodegeneration: where are we now?, J. Neurochem., 97, 1634, 10.1111/j.1471-4159.2006.03907.x

Alzheimer, 1995, An English translation of Alzheimer's 1907 paper, "Uber eine eigenartige Erkankung der Hirnrinde", Clin. Anat., 8, 429, 10.1002/ca.980080612

Goedert, 2006, A century of Alzheimer's disease, Science, 314, 777, 10.1126/science.1132814

M. Prince, A. Wimo, M. Guerchet, G. Ali, Y. Wu, M. Prina, World Alzheimer Report 2015. The global impact of dementia. An analysis of prevalence, incidence, cost and trends, Alzheimer’s Disease International, London, 2015.

B. Duthey, Background paper 6.11: Alzheimer disease and other dementias, Priority Medicines for Europe and the World. "A public Health Approach to Innovation", 2004, pp. 1–74.

Mattson, 2004, Pathways towards and away from Alzheimer's disease, Nature, 430, 631, 10.1038/nature02621

Soto-Rojas, 2015, Neuroinflammation and alteration of the blood-brain barrier in Alzheimers disease

Janicki, 2010, Hormonal influences on cognition and risk for Alzheimer disease, Curr. Neurol. Neurosci. Rep., 10, 359, 10.1007/s11910-010-0122-6

Glenner, 1984, Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein, Biochem. Biophys. Res. Commun., 120, 885, 10.1016/S0006-291X(84)80190-4

Minati, 2009, Current concepts in Alzheimer's disease: a multidisciplinary review, Am. J. Alzheimer'S. Dis. Other Dement., 24, 95, 10.1177/1533317508328602

Goedert, 2015, NEURODEGENERATION. Alzheimer's and Parkinson's diseases: the prion concept in relation to assembled Abeta, tau, and alpha-synuclein, Science, 349, 1255555, 10.1126/science.1255555

Grundke-Iqbal, 1986, Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology, Proc. Natl. Acad. Sci. USA, 83, 4913, 10.1073/pnas.83.13.4913

Giraldo, 2014, Abeta and tau toxicities in Alzheimer's are linked via oxidative stress-induced p38 activation: protective role of vitamin E, Redox Biol., 2, 873, 10.1016/j.redox.2014.03.002

Jakob‐Roetne, 2009, Alzheimer's disease: from pathology to therapeutic approaches, Angew. Chem. Int. Ed., 48, 3030, 10.1002/anie.200802808

Nalivaeva, 2013, The amyloid precursor protein: a biochemical enigma in brain development, function and disease, FEBS Lett., 587, 2046, 10.1016/j.febslet.2013.05.010

Chow, 2010, An overview of APP processing enzymes and products, NeuroMol. Med., 12, 1, 10.1007/s12017-009-8104-z

Willem, 2015, eta-Secretase processing of APP inhibits neuronal activity in the hippocampus, Nature, 526, 443, 10.1038/nature14864

Haass, 1992, Amyloid β-peptide is produced by cultured cells during normal metabolism, Nature, 359, 322, 10.1038/359322a0

Saido, 2012, Proteolytic degradation of amyloid beta-protein, Cold Spring Harb. Perspect. Med., 2, a006379, 10.1101/cshperspect.a006379

Puzzo, 2013, Amyloid-beta peptide: Dr. Jekyll or Mr. Hyde?, J. Alzheimers Dis., 33, S111

Barber, 2012, The genetics of Alzheimer's disease, Scientifica (Cairo), 2012, 246210

Wiseman, 2015, A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome, Nat. Rev. Neurosci., 10.1038/nrn3983

Nasica-Labouze, 2015, Amyloid β protein and Alzheimer’s disease: when computer simulations complement experimental studies, Chem. Rev., 115, 3518, 10.1021/cr500638n

Cruts, 2012, Locus-specific mutation databases for neurodegenerative brain diseases, Human. Mutat., 33, 1340, 10.1002/humu.22117

Karran, 2011, The amyloid cascade hypothesis for Alzheimer's disease: an appraisal for the development of therapeutics, Nat. Rev. Drug Discov., 10, 698, 10.1038/nrd3505

Holtzman, 2011, Alzheimer’s disease: the challenge of the second century, Sci. Transl. Med., 3, 10.1126/scitranslmed.3002369

Hardy, 1991, Amyloid deposition as the central event in the aetiology of Alzheimer's disease, Trends Pharmacol. Sci., 12, 383, 10.1016/0165-6147(91)90609-V

Selkoe, 1991, The molecular pathology of Alzheimer's disease, Neuron, 6, 487, 10.1016/0896-6273(91)90052-2

Beyreuther, 1991, Amyloid precursor protein (APP) and beta A4 amyloid in the etiology of Alzheimer's disease: precursor-product relationships in the derangement of neuronal function, Brain Pathol., 1, 241, 10.1111/j.1750-3639.1991.tb00667.x

Hardy, 1992, Alzheimer's disease: the amyloid cascade hypothesis, Science, 256, 184, 10.1126/science.1566067

Selkoe, 2016, The amyloid hypothesis of Alzheimer's disease at 25 years, EMBO Mol. Med., 8, 595, 10.15252/emmm.201606210

Reitz, 2012, Alzheimer's disease and the amyloid cascade hypothesis: a critical review, Int. J. Alzheimer’s Dis., 2012

Herrup, 2015, The case for rejecting the amyloid cascade hypothesis, Nat. Neurosci., 794, 10.1038/nn.4017

Ding, 2003, Mechanism for the α-helix to β-hairpin transition, Protein.: Struct. Funct. Bioinforma., 53, 220, 10.1002/prot.10468

Pham, 2010, Progressive accumulation of amyloid-beta oligomers in Alzheimer's disease and in amyloid precursor protein transgenic mice is accompanied by selective alterations in synaptic scaffold proteins, FEBS J., 277, 3051, 10.1111/j.1742-4658.2010.07719.x

Forloni, 2016, Oligomeropathies and pathogenesis of Alzheimer and Parkinson's diseases, Mov. Disord., 10.1002/mds.26624

Deshpande, 2006, Different conformations of amyloid β induce neurotoxicity by distinct mechanisms in human cortical neurons, J. Neurosci., 26, 6011, 10.1523/JNEUROSCI.1189-06.2006

Glabe, 2006, Common mechanisms of amyloid oligomer pathogenesis in degenerative disease, Neurobiol. Aging, 27, 570, 10.1016/j.neurobiolaging.2005.04.017

Lovell, 1998, Copper, iron and zinc in Alzheimer's disease senile plaques, J. Neurol. Sci., 158, 47, 10.1016/S0022-510X(98)00092-6

Tiiman, 2013, The missing link in the amyloid cascade of Alzheimer’s disease – metal ions, Neurochem. Int., 62, 367, 10.1016/j.neuint.2013.01.023

Faller, 2013, Role of metal ions in the self-assembly of the Alzheimer’s amyloid-β peptide, Inorg. Chem., 52, 12193, 10.1021/ic4003059

Bayir, 2005, Reactive oxygen species, Crit. Care Med., 33, S498, 10.1097/01.CCM.0000186787.64500.12

Butterfield, 2010, Involvements of the lipid peroxidation product, HNE, in the pathogenesis and progression of Alzheimer's disease, Biochim. Biophys. Acta, 1801, 924, 10.1016/j.bbalip.2010.02.005

Wang, 2014, Oxidative stress and mitochondrial dysfunction in Alzheimer's disease, Biochim. Biophys. Acta, 1842, 1240, 10.1016/j.bbadis.2013.10.015

Butterfield, 2002, Lipid peroxidation and protein oxidation in Alzheimer's disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress, Free Radic. Biol. Med., 32, 1050, 10.1016/S0891-5849(02)00794-3

Sultana, 2006, Redox proteomics identification of oxidized proteins in Alzheimer's disease hippocampus and cerebellum: an approach to understand pathological and biochemical alterations in AD, Neurobiol. Aging, 27, 1564, 10.1016/j.neurobiolaging.2005.09.021

Hensley, 1995, Brain regional correspondence between Alzheimer's disease histopathology and biomarkers of protein oxidation, J. Neurochem., 65, 2146, 10.1046/j.1471-4159.1995.65052146.x

Butterfield, 1999, Amyloid beta-peptide-associated free radical oxidative stress, neurotoxicity, and Alzheimer's disease, Methods Enzymol., 309, 746, 10.1016/S0076-6879(99)09050-3

Butterfield, 2014, The 2013 SFRBM discovery award: selected discoveries from the butterfield laboratory of oxidative stress and its sequela in brain in cognitive disorders exemplified by Alzheimer disease and chemotherapy induced cognitive impairment, Free Radic. Biol. Med., 74, 157, 10.1016/j.freeradbiomed.2014.06.006

Zhao, 2013, Oxidative stress and the pathogenesis of Alzheimer's disease, Oxid. Med. Cell. Longev., 2013, 316523, 10.1155/2013/316523

Selfridge, 2013, Role of mitochondrial homeostasis and dynamics in Alzheimer's disease, Neurobiol. Dis., 51, 3, 10.1016/j.nbd.2011.12.057

Butterfield, 2007, Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment, Free Radic. Biol. Med., 43, 658, 10.1016/j.freeradbiomed.2007.05.037

Granold, 2015, High membrane protein oxidation in the human cerebral cortex, Redox Biol., 4, 200, 10.1016/j.redox.2014.12.013

Gow, 1996, Effects of peroxynitrite-induced protein modifications on tyrosine phosphorylation and degradation, FEBS Lett., 385, 63, 10.1016/0014-5793(96)00347-X

Markesbery, 1998, Four-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimer's disease, Neurobiol. Aging, 19, 33, 10.1016/S0197-4580(98)00009-8

Hardas, 2013, Oxidative modification of lipoic acid by HNE in Alzheimer disease brain, Redox Biol., 1, 80, 10.1016/j.redox.2013.01.002

Sayre, 1997, 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer's disease, J. Neurochem., 68, 2092, 10.1046/j.1471-4159.1997.68052092.x

Mecocci, 1994, Oxidative damage to mitochondrial DNA is increased in Alzheimer's disease, Ann. Neurol., 36, 747, 10.1002/ana.410360510

Gabbita, 1998, Increased nuclear DNA oxidation in the brain in Alzheimer's disease, J. Neurochem., 71, 2034, 10.1046/j.1471-4159.1998.71052034.x

Lovell, 2011, Oxidatively modified nucleic acids in preclinical Alzheimer's disease (PCAD) brain, Mech. Ageing Dev., 132, 443, 10.1016/j.mad.2011.08.003

Coppede, 2015, DNA damage in neurodegenerative diseases, Mutat. Res., 776, 84, 10.1016/j.mrfmmm.2014.11.010

Shan, 2006, Quantification of oxidized RNAs in Alzheimer's disease, Neurobiol. Aging, 27, 657, 10.1016/j.neurobiolaging.2005.03.022

Butterfield, 2010, Amyloidogenic protein-membrane interactions: mechanistic insight from model systems, Angew. Chem. Int. Ed. Engl., 49, 5628, 10.1002/anie.200906670

Castegna, 2004, Modulation of phospholipid asymmetry in synaptosomal membranes by the lipid peroxidation products, 4-hydroxynonenal and acrolein: implications for Alzheimer's disease, Brain Res., 1004, 193, 10.1016/j.brainres.2004.01.036

Subramaniam, 1997, The lipid peroxidation product, 4-hydroxy-2-trans-nonenal, alters the conformation of cortical synaptosomal membrane proteins, J. Neurochem., 69, 1161, 10.1046/j.1471-4159.1997.69031161.x

Tramutola, 2016, Oxidative stress, protein modification and Alzheimer disease, Brain Res. Bull.

Aluise, 2011, Redox proteomics analysis of brains from subjects with amnestic mild cognitive impairment compared to brains from subjects with preclinical Alzheimer's disease: insights into memory loss in MCI, J. Alzheimers Dis. : JAD, 23, 257, 10.3233/JAD-2010-101083

Perluigi, 2009, Redox proteomics identification of 4-hydroxynonenal-modified brain proteins in Alzheimer's disease: role of lipid peroxidation in Alzheimer's disease pathogenesis, Proteom. Clin. Appl., 3, 682, 10.1002/prca.200800161

Reed, 2008, Redox proteomic identification of 4-hydroxy-2-nonenal-modified brain proteins in amnestic mild cognitive impairment: insight into the role of lipid peroxidation in the progression and pathogenesis of Alzheimer's disease, Neurobiol. Dis., 30, 107, 10.1016/j.nbd.2007.12.007

Terni, 2010, Mitochondrial ATP-synthase in the entorhinal cortex is a target of oxidative stress at stages I/II of Alzheimer's disease pathology, Brain Pathol., 20, 222, 10.1111/j.1750-3639.2009.00266.x

Schagger, 1995, Human diseases with defects in oxidative phosphorylation. 2. F1F0 ATP-synthase defects in Alzheimer disease revealed by blue native polyacrylamide gel electrophoresis, Eur. J. Biochem., 227, 916, 10.1111/j.1432-1033.1995.tb20219.x

Cha, 2015, Mitochondrial ATP synthase activity is impaired by suppressed O-GlcNAcylation in Alzheimer's disease, Hum. Mol. Genet., 24, 6492, 10.1093/hmg/ddv358

Chen, 2013, Decoding Alzheimer's disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies, Prog. Neurobiol., 108, 21, 10.1016/j.pneurobio.2013.06.004

Butterfield, 2014, Elevated risk of type 2 diabetes for development of Alzheimer disease: a key role for oxidative stress in brain, Biochim. Biophys. Acta, 1842, 1693, 10.1016/j.bbadis.2014.06.010

Di Domenico, 2016, The triangle of death in Alzheimer's disease brain: the aberrant cross-talk among energy metabolism, mammalian target of rapamycin signaling, and protein homeostasis revealed by redox proteomics, Antioxid. Redox Signal.

Barone, 2016, Impairment of biliverdin reductase-A promotes brain insulin resistance in Alzheimer disease: a new paradigm, Free Radic. Biol. Med., 91, 127, 10.1016/j.freeradbiomed.2015.12.012

Ito, 2007, Cerebral clearance of human amyloid-beta peptide (1-40) across the blood-brain barrier is reduced by self-aggregation and formation of low-density lipoprotein receptor-related protein-1 ligand complexes, J. Neurochem., 103, 2482, 10.1111/j.1471-4159.2007.04938.x

Sagare, 2007, Clearance of amyloid-beta by circulating lipoprotein receptors, Nat. Med., 13, 1029, 10.1038/nm1635

Jeynes, 2008, Evidence for altered LRP/RAGE expression in Alzheimer lesion pathogenesis, Curr. Alzheimer Res., 5, 432, 10.2174/156720508785908937

Owen, 2010, Oxidative modification to LDL receptor-related protein 1 in hippocampus from subjects with Alzheimer disease: implications for Abeta accumulation in AD brain, Free Radic. Biol. Med., 49, 1798, 10.1016/j.freeradbiomed.2010.09.013

Liu, 2005, Alzheimer-specific epitopes of tau represent lipid peroxidation-induced conformations, Free Radic. Biol. Med., 38, 746, 10.1016/j.freeradbiomed.2004.11.005

Horiguchi, 2003, Nitration of tau protein is linked to neurodegeneration in tauopathies, Am. J. Pathol., 163, 1021, 10.1016/S0002-9440(10)63462-1

Violet, 2014, A major role for Tau in neuronal DNA and RNA protection in vivo under physiological and hyperthermic conditions, Front. Cell. Neurosci., 8, 84, 10.3389/fncel.2014.00084

Coppede, 2010, Evidence linking genetics, environment, and epigenetics to impaired DNA repair in Alzheimer's disease, J. Alzheimer's Dis. : JAD, 20, 953, 10.3233/JAD-2010-1415

Weissman, 2007, Defective DNA base excision repair in brain from individuals with Alzheimer's disease and amnestic mild cognitive impairment, Nucleic Acids Res., 35, 5545, 10.1093/nar/gkm605

Su, 1997, Neuronal DNA damage precedes tangle formation and is associated with up-regulation of nitrotyrosine in Alzheimer's disease brain, Brain Res., 774, 193, 10.1016/S0006-8993(97)81703-9

Beel, 2010, Direct binding of cholesterol to the amyloid precursor protein: an important interaction in lipid-Alzheimer's disease relationships?, Biochim. Biophys. Acta, 1801, 975, 10.1016/j.bbalip.2010.03.008

Puglielli, 2001, Acyl-coenzyme A: cholesterol acyltransferase modulates the generation of the amyloid beta-peptide, Nat. Cell Biol., 3, 905, 10.1038/ncb1001-905

Bjorkhem, 2009, Oxysterols and neurodegenerative diseases, Mol. Asp. Med., 30, 171, 10.1016/j.mam.2009.02.001

Usui, 2009, Site-specific modification of Alzheimer's peptides by cholesterol oxidation products enhances aggregation energetics and neurotoxicity, Proc. Natl. Acad. Sci. USA, 106, 18563, 10.1073/pnas.0804758106

Murray, 2007, Membrane-mediated amyloidogenesis and the promotion of oxidative lipid damage by amyloid beta proteins, J. Biol. Chem., 282, 9335, 10.1074/jbc.M608589200

Zhang, 2004, Metabolite-initiated protein misfolding may trigger Alzheimer's disease, Proc. Natl. Acad. Sci. USA, 101, 4752, 10.1073/pnas.0400924101

Klein, 2004, Small assemblies of unmodified amyloid beta-protein are the proximate neurotoxin in Alzheimer's disease, Neurobiol. Aging, 25, 569, 10.1016/j.neurobiolaging.2004.02.010

Bjorkhem, 2006, Crossing the barrier: oxysterols as cholesterol transporters and metabolic modulators in the brain, J. Intern. Med., 260, 493, 10.1111/j.1365-2796.2006.01725.x

Abildayeva, 2006, 24(S)-hydroxycholesterol participates in a liver X receptor-controlled pathway in astrocytes that regulates apolipoprotein E-mediated cholesterol efflux, J. Biol. Chem., 281, 12799, 10.1074/jbc.M601019200

Heverin, 2005, Crossing the barrier: net flux of 27-hydroxycholesterol into the human brain, J. Lipid Res., 46, 1047, 10.1194/jlr.M500024-JLR200

Gamba, 2012, The link between altered cholesterol metabolism and Alzheimer's disease, Ann. N. Y. Acad. Sci., 1259, 54, 10.1111/j.1749-6632.2012.06513.x

Heverin, 2004, Changes in the levels of cerebral and extracerebral sterols in the brain of patients with Alzheimer's disease, J. Lipid Res., 45, 186, 10.1194/jlr.M300320-JLR200

Prasanthi, 2009, Differential effects of 24-hydroxycholesterol and 27-hydroxycholesterol on beta-amyloid precursor protein levels and processing in human neuroblastoma SH-SY5Y cells, Mol. Neurodegener., 4, 1, 10.1186/1750-1326-4-1

Gamba, 2014, Up-regulation of beta-amyloidogenesis in neuron-like human cells by both 24- and 27-hydroxycholesterol: protective effect of N-acetyl-cysteine, Aging Cell, 13, 561, 10.1111/acel.12206

Leoni, 2011, Oxysterols as biomarkers in neurodegenerative diseases, Chem. Phys. Lipids, 164, 515, 10.1016/j.chemphyslip.2011.04.002

Testa, 2016, Changes in brain oxysterols at different stages of Alzheimer's disease: their involvement in neuroinflammation, Redox Biol., 10, 24, 10.1016/j.redox.2016.09.001

Chico, 2013, Oxidative stress and APO E polymorphisms in Alzheimer's disease and in mild cognitive impairment, Free Radic. Res., 47, 569, 10.3109/10715762.2013.804622

Fernandes, 1999, Influence of apolipoprotein E genotype on blood redox status of Alzheimer's disease patients, Int. J. Mol. Med., 4, 179

Jenner, 2010, The effect of APOE genotype on brain levels of oxysterols in young and old human APOE epsilon2, epsilon3 and epsilon4 knock-in mice, Neuroscience, 169, 109, 10.1016/j.neuroscience.2010.04.026

Di Domenico, 2016, Oxidative signature of cerebrospinal fluid from mild cognitive impairment and Alzheimer disease patients, Free Radic. Biol. Med., 91, 1, 10.1016/j.freeradbiomed.2015.12.004

Faller, 2012, A bioinorganic view of Alzheimer's disease: when misplaced metal ions (re)direct the electrons to the wrong target, Chemistry, 18, 15910, 10.1002/chem.201202697

Lutsenko, 2010, Copper handling machinery of the brain, Metallomics : Integr. Biomet. Sci., 2, 596, 10.1039/c0mt00006j

Delangle, 2012, Chelation therapy in Wilson's disease: from D-penicillamine to the design of selective bioinspired intracellular Cu(I) chelators, Dalton Trans., 41, 6359, 10.1039/c2dt12188c

Barnham, 2014, Biological metals and metal-targeting compounds in major neurodegenerative diseases, Chem. Soc. Rev., 43, 6727, 10.1039/C4CS00138A

Bonda, 2011, Role of metal dyshomeostasis in Alzheimer's disease, Metallomics : Integr. Biomet. Sci., 3, 267, 10.1039/c0mt00074d

Miller, 2006, Synchrotron-based infrared and X-ray imaging shows focalized accumulation of Cu and Zn co-localized with beta-amyloid deposits in Alzheimer's disease, J. Struct. Biol., 155, 30, 10.1016/j.jsb.2005.09.004

Leskovjan, 2009, Amyloid plaques in PSAPP mice bind less metal than plaques in human Alzheimer's disease, Neuroimage, 47, 1215, 10.1016/j.neuroimage.2009.05.063

Duce, 2010, Iron-export ferroxidase activity of beta-amyloid precursor protein is inhibited by zinc in Alzheimer's disease, Cell, 142, 857, 10.1016/j.cell.2010.08.014

Rogers, 2002, An iron-responsive element type II in the 5'-untranslated region of the Alzheimer's amyloid precursor protein transcript, J. Biol. Chem., 277, 45518, 10.1074/jbc.M207435200

Bellingham, 2004, Copper depletion down-regulates expression of the Alzheimer's disease amyloid-beta precursor protein gene, J. Biol. Chem., 279, 20378, 10.1074/jbc.M400805200

Dahms, 2012, Metal binding dictates conformation and function of the amyloid precursor protein (APP) E2 domain, J. Mol. Biol., 416, 438, 10.1016/j.jmb.2011.12.057

Multhaup, 1996, The amyloid precursor protein of Alzheimer's disease in the reduction of copper(II) to copper(I), Science, 271, 1406, 10.1126/science.271.5254.1406

James, 2012, Elevated labile Cu is associated with oxidative pathology in Alzheimer disease, Free Radic. Biol. Med., 52, 298, 10.1016/j.freeradbiomed.2011.10.446

Kozlowski, 2012, Copper, zinc and iron in neurodegenerative diseases (Alzheimer's, Parkinson's and prion diseases), Coord. Chem. Rev., 256, 2129, 10.1016/j.ccr.2012.03.013

Migliorini, 2012, Structural characterization of Cu 2+, Ni 2+ and Zn 2+ binding sites of model peptides associated with neurodegenerative diseases, Coord. Chem. Rev., 256, 352, 10.1016/j.ccr.2011.07.004

Tõugu, 2012, Coordination of zinc ions to the key proteins of neurodegenerative diseases: aβ, APP, α-synuclein and PrP, Coord. Chem. Rev., 256, 2219, 10.1016/j.ccr.2011.12.008

Zirah, 2006, Structural changes of region 1-16 of the Alzheimer disease amyloid beta-peptide upon zinc binding and in vitro aging, J. Biol. Chem., 281, 2151, 10.1074/jbc.M504454200

Alies, 2016, Zinc (II) binding site to the amyloid-β peptide: insights from spectroscopic studies with a wide series of modified peptides, Inorg. Chem., 55, 10499, 10.1021/acs.inorgchem.6b01733

Noel, 2014, Use of a new water-soluble Zn sensor to determine Zn affinity for the amyloid-beta peptide and relevant mutants, Metallomics : Integr. Biomet. Sci., 6, 1220, 10.1039/c4mt00016a

Talmard, 2007, Zinc binding to amyloid-beta: isothermal titration calorimetry and Zn competition experiments with Zn sensors, Biochemistry, 46, 13658, 10.1021/bi701355j

Hureau, 2012, Coordination of redox active metal ions to the amyloid precursor protein and to amyloid-β peptides involved in Alzheimer disease. Part 2: dependence of Cu(II) binding sites with Aβ sequences, Coord. Chem. Rev., 256, 2175, 10.1016/j.ccr.2012.03.034

Hureau, 2012, Coordination of redox active metal ions to the amyloid precursor protein and to amyloid-β peptides involved in Alzheimer disease. Part 1: an overview, Coord. Chem. Rev., 256, 2164, 10.1016/j.ccr.2012.03.037

Zawisza, 2012, Affinity of copper and zinc ions to proteins and peptides related to neurodegenerative conditions (Aβ, APP, α-synuclein, PrP), Coord. Chem. Rev., 256, 2297, 10.1016/j.ccr.2012.03.012

Dorlet, 2009, Pulse EPR Spectroscopy Reveals The Coordination Sphere Of Copper(II) ions in the 1–16 amyloid-β peptide: a key role of the first two n-terminus residues, Angew. Chem. Int. Ed., 48, 9273, 10.1002/anie.200904567

Hureau, 2009, Deprotonation of the Asp1- Ala2 peptide bond induces modification of the dynamic copper (II) environment in the amyloid‐β peptide near physiological pH, Angew. Chem. Int. Ed., 48, 9522, 10.1002/anie.200904512

Drew, 2009, Alanine-2 carbonyl is an oxygen ligand in Cu2+ coordination of Alzheimer’s disease amyloid-β peptide− relevance to N-terminally truncated forms, J. Am. Chem. Soc., 131, 8760, 10.1021/ja903669a

Drew, 2011, The heterogeneous nature of Cu2+ interactions with Alzheimer’s amyloid-β peptide, Acc. Chem. Res., 44, 1146, 10.1021/ar200014u

Drew, 2009, Pleomorphic copper coordination by Alzheimer’s disease amyloid-β peptide, J. Am. Chem. Soc., 131, 1195, 10.1021/ja808073b

Alies, 2011, pH-dependent Cu(II) coordination to amyloid-β peptide: impact of sequence alterations, including the H6R and D7N familial mutations, Inorg. Chem., 50, 11192, 10.1021/ic201739n

Arena, 2012, Copper(II) interaction with amyloid-β: affinity and speciation, Coord. Chem. Rev., 256, 3, 10.1016/j.ccr.2011.07.012

Alies, 2013, Cu(II) affinity for the Alzheimer's peptide: tyrosine fluorescence studies revisited, Anal. Chem., 85, 1501, 10.1021/ac302629u

Conte-Daban, 2017, Link between affinity and cu(ii) binding sites to amyloid-beta peptides evaluated by a new water-soluble UV–visible ratiometric dye with a moderate Cu(II) affinity, Anal. Chem., 89, 2155, 10.1021/acs.analchem.6b04979

Shearer, 2008, The amyloid-beta peptide of Alzheimer's disease binds Cu(I) in a linear bis-his coordination environment: insight into a possible neuroprotective mechanism for the amyloid-beta peptide, J. Am. Chem. Soc., 130, 17826, 10.1021/ja805940m

Himes, 2007, Synthesis and X-ray absorption spectroscopy structural studies of Cu (I) complexes of histidylhistidine peptides: the predominance of linear 2-coordinate geometry, J. Am. Chem. Soc., 129, 5352, 10.1021/ja0708013

Himes, 2008, Structural studies of copper (i) complexes of amyloid‐β peptide fragments: formation of two‐coordinate bis (histidine) complexes, Angew. Chem. Int. Ed., 47, 9084, 10.1002/anie.200803908

Lu, 2010, Copper(i) and copper(ii) binding to β-amyloid 16 (Aβ16) studied by electrospray ionization mass spectrometry, Metallomics, 2, 10.1039/c004693k

Alies, 2012, Reevaluation of copper(i) affinity for amyloid-β peptides by competition with ferrozine—an unusual copper(i) indicator, Chem. – Eur. J., 18, 1161, 10.1002/chem.201102746

Feaga, 2011, Affinity of Cu+ for the copper-binding domain of the amyloid-β peptide of Alzheimer’s disease, Inorg. Chem., 50, 1614, 10.1021/ic100967s

Young, 2014, An integrated study of the affinities of the Aβ16 peptide for Cu (I) and Cu (II): implications for the catalytic production of reactive oxygen species, Metallomics, 6, 505, 10.1039/C4MT00001C

Valensin, 2011, Exploring the reactions of beta-amyloid (Abeta) peptide 1-28 with Al(III) and Fe(III) ions, Inorg. Chem., 50, 6865, 10.1021/ic201069v

Bousejra-ElGarah, 2011, Iron(II) binding to amyloid-β, the Alzheimer’s peptide, Inorg. Chem., 50, 9024, 10.1021/ic201233b

Halliwell, 2006, Oxidative stress and neurodegeneration: where are we now?, J. Neurochem., 97, 1634, 10.1111/j.1471-4159.2006.03907.x

Nakamura, 2007, Three histidine residues of amyloid-β peptide control the redox activity of copper and iron, Biochemistry, 46, 12737, 10.1021/bi701079z

Smith, 1997, Iron accumulation in Alzheimer disease is a source of redox-generated free radicals, Proc. Natl. Acad. Sci. USA, 94, 9866, 10.1073/pnas.94.18.9866

Guilloreau, 2007, Redox chemistry of copper–amyloid-β: the generation of hydroxyl radical in the presence of ascorbate is linked to redox-potentials and aggregation state, ChemBioChem, 8, 1317, 10.1002/cbic.200700111

Nadal, 2008, Amyloid β–Cu2+ complexes in both monomeric and fibrillar forms do not generate H2O2 catalytically but quench hydroxyl radicals, Biochemistry, 47, 11653, 10.1021/bi8011093

Baruch-Suchodolsky, 2008, Soluble amyloid β1-28-copper(I)/copper(II)/iron(II) complexes are potent antioxidants in cell-free systems, Biochemistry, 47, 7796, 10.1021/bi800114g

Hureau, 2009, Aβ-mediated ROS production by Cu ions: structural insights, mechanisms and relevance to Alzheimer's disease, Biochimie, 91, 1212, 10.1016/j.biochi.2009.03.013

Dikalov, 2004, Cupric–amyloid β peptide complex stimulates oxidation of ascorbate and generation of hydroxyl radical, Free Radic. Biol. Med., 36, 340, 10.1016/j.freeradbiomed.2003.11.004

Reybier, 2016, Free superoxide is an intermediate in the production of H2O2 by copper(I)-abeta peptide and O2, Angew. Chem. Int. Ed. Engl., 55, 1085, 10.1002/anie.201508597

Balland, 2010, Electrochemical and homogeneous electron transfers to the Alzheimer amyloid-beta copper complex follow a preorganization mechanism, Proc. Natl. Acad. Sci. USA, 107, 17113, 10.1073/pnas.1011315107

La Penna, 2013, Identifying, By first-principles simulations, Cu[amyloid-β] species making fenton-type reactions in Alzheimer’s disease, J. Phys. Chem. B, 117, 16455, 10.1021/jp410046w

Mirats, 2015, Dioxygen activation in the Cu–amyloid β complex, Phys. Chem. Chem. Phys., 17, 27270, 10.1039/C5CP04025F

Furlan, 2012, Modeling copper binding to the amyloid-β peptide at different pH: toward a molecular mechanism for Cu reduction, J. Phys. Chem. B, 116, 11899, 10.1021/jp308977s

Prosdocimi, 2016, On the generation of OH·radical species from H2O2 by Cu (I) amyloid beta peptide model complexes: a DFT investigation, J. Biol. Inorg. Chem., 21, 197, 10.1007/s00775-015-1322-y

Cassagnes, 2013, The Catalytically Active Copper-Amyloid-Beta State: coordination Site Responsible for Reactive Oxygen Species Production, Angew. Chem. Int. Ed., 52, 11110, 10.1002/anie.201305372

Cheignon, 2017, Real-time evolution of Aβ40 metal-catalyzed oxidation reveals Asp1 as the main target and a dependence on metal binding site, Inorg. Chim. Acta

Cheignon, 2017, Identification of key structural features of the elusive Cu-A[small beta] complex that generates ROS in Alzheimer's disease, Chem. Sci., 8, 5107, 10.1039/C7SC00809K

Devasagayam, 2004, Free radicals and antioxidants in human health: current status and future prospects, JAPI, 52, 4

Gardès-Albert, 2005, Aspects physicochimiques des radicaux libres centrés sur l'oxygène, 6

Bielski, 1985, Reactivity of HO2/O–2 radicals in aqueous solution, J. Phys. Chem. Ref. Data, 14, 1041, 10.1063/1.555739

L.M. Dorfman, G.E. Adams, Reactivity of the hydroxyl radical in aqueous solutions, DTIC Document, 1973.

Wardman, 1989, Reduction potentials of one-electron couples involving free radicals in aqueous solution, J. Phys. Chem. Ref. Data, 18, 1637, 10.1063/1.555843

Markesbery, 1997, Oxidative stress hypothesis in Alzheimer's disease, Free Radic. Biol. Med., 23, 134, 10.1016/S0891-5849(96)00629-6

Butterfield, 2001, Evidence of oxidative damage in Alzheimer's disease brain: central role for amyloid β-peptide, Trends Mol. Med., 7, 548, 10.1016/S1471-4914(01)02173-6

Näslund, 1994, Relative abundance of Alzheimer A beta amyloid peptide variants in Alzheimer disease and normal aging, Proc. Natl. Acad. Sci. USA, 91, 8378, 10.1073/pnas.91.18.8378

Stadtman, 2003, Free radical-mediated oxidation of free amino acids and amino acid residues in proteins, Amino Acids, 25, 207, 10.1007/s00726-003-0011-2

Stadtman, 1993, Oxidation of free amino acids and amino acid residues in proteins by radiolysis and by metal-catalyzed reactions, Annu. Rev. Biochem., 62, 797, 10.1146/annurev.bi.62.070193.004053

Stadtman, 1995, Role of oxidized amino acids in protein breakdown and stability, Methods Enzymol., 258, 379, 10.1016/0076-6879(95)58057-3

Bonnefont-Rousselot, 2005, Oxydation des acides aminés et des protéines, 147

Inoue, 2009, Screening assay for metal-catalyzed oxidation inhibitors using liquid chromatography–mass spectrometry with an n-terminal β-amyloid peptide, Anal. Chem., 81, 1819, 10.1021/ac802162n

Kowalik-Jankowska, 2004, Products of Cu(II)-catalyzed oxidation in the presence of hydrogen peroxide of the 1–10, 1–16 fragments of human and mouse β-amyloid peptide, J. Inorg. Biochem., 98, 940, 10.1016/j.jinorgbio.2004.03.001

Schöneich, 2002, Cu(II)-catalyzed oxidation of β-amyloid peptide targets His13 and His14 over His6: detection of 2-Oxo-histidine by HPLC-MS/MS, Chem. Res. Toxicol., 15, 717, 10.1021/tx025504k

Inoue, 2006, Liquid chromatography/tandem mass spectrometry characterization of oxidized amyloid beta peptides as potential biomarkers of Alzheimer's disease, Rapid Commun. Mass Spectrom., 20, 911, 10.1002/rcm.2395

Schöneich, 2000, Mechanisms of metal-catalyzed oxidation of histidine to 2-oxo-histidine in peptides and proteins, J. Pharm. Biomed. Anal., 21, 1093, 10.1016/S0731-7085(99)00182-X

Schoneich, 2004, Selective Cu2+/ascorbate-dependent oxidation of alzheimer's disease beta-amyloid peptides, Ann. N. Y. Acad. Sci., 1012, 164, 10.1196/annals.1306.013

Taniguchi, 2014, Attenuation of the aggregation and neurotoxicity of amyloid-beta peptides by catalytic photooxygenation, Angew. Chem., 53, 1382, 10.1002/anie.201308001

Atwood, 2004, Copper mediates dityrosine cross-linking of Alzheimer's amyloid-β, Biochemistry, 43, 560, 10.1021/bi0358824

Gunn, 2012, Rapid generation of dityrosine cross-linked Aβ oligomers via Cu-redox cycling, 3

Al-Hilaly, 2013, A central role for dityrosine crosslinking of Amyloid-β in Alzheimer’s disease, Acta Neuropathol. Commun., 1, 10.1186/2051-5960-1-83

Barnham, 2004, Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer's disease beta-amyloid, FASEB J., 18, 1427, 10.1096/fj.04-1890fje

Moskovitz, 2014, Methionine sulfoxide reductase system in health and disease, Austin J. Pharmacol. Ther., 2, 3

Schöneich, 2003, Free radical reactions of methionine in peptides: mechanisms relevant to β-amyloid oxidation and Alzheimer's disease, J. Am. Chem. Soc., 125, 13700, 10.1021/ja036733b

Schöneich, 2005, Methionine oxidation by reactive oxygen species: reaction mechanisms and relevance to Alzheimer's disease, Biochim Biophys. Acta, 1703, 111, 10.1016/j.bbapap.2004.09.009

Butterfield, 2011, Methionine-35 of aβ (1–42): importance for oxidative stress in Alzheimer disease, J. Amino Acids, 2011, 10.4061/2011/198430

Ali, 2005, Methionine regulates copper/hydrogen peroxide oxidation products of Aβ, J. Pept. Sci., 11, 353, 10.1002/psc.626

Kanski, 2002, Role of glycine-33 and methionine-35 in Alzheimer’s amyloid β-peptide 1–42-associated oxidative stress and neurotoxicity, Biochimica et Biophysica Acta, Mol. Basis Dis., 1586, 190, 10.1016/S0925-4439(01)00097-7

Slemmon, 2012, Measurement of Abeta1-42 in cerebrospinal fluid is influenced by matrix effects, J. Neurochem., 120, 325, 10.1111/j.1471-4159.2011.07553.x

Perret-Liaudet, 2012, Risk of Alzheimer's disease biological misdiagnosis linked to cerebrospinal collection tubes, J. Alzheimer's Dis. : JAD, 31, 13, 10.3233/JAD-2012-120361

Bros, 2015, Quantitative detection of amyloid-beta peptides by mass spectrometry: state of the art and clinical applications, Clin. Chem. Lab. Med., 53, 1483, 10.1515/cclm-2014-1048

Rogeberg, 2015, Identification of amyloid beta mid-domain fragments in human cerebrospinal fluid, Biochimie, 113, 86, 10.1016/j.biochi.2015.03.022

Lesne, 2013, Brain amyloid-beta oligomers in ageing and Alzheimer's disease, Brain, 136, 1383, 10.1093/brain/awt062

Youmans, 2012, Intraneuronal Abeta detection in 5xFAD mice by a new Abeta-specific antibody, Mol. Neurodegener., 7, 8, 10.1186/1750-1326-7-8

Naslund, 1994, Relative abundance of Alzheimer A beta amyloid peptide variants in Alzheimer disease and normal aging, Proc. Natl. Acad. Sci. USA, 91, 8378, 10.1073/pnas.91.18.8378

Kim, 1988, Production and characterization of monoclonal antibodies reactive to synthetic cerebrovascular amyloid peptide, Neurosci. Res. Commun., 2, 121

Portelius, 2006, Determination of beta-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation-mass spectrometry, J. Proteome Res., 5, 1010, 10.1021/pr050475v

Butterfield, 2010, In vivo oxidative stress in brain of Alzheimer disease transgenic mice: requirement for methionine 35 in amyloid beta-peptide of APP, Free Radic. Biol. Med., 48, 136, 10.1016/j.freeradbiomed.2009.10.035

Inoue, 2006, Liquid chromatography/tandem mass spectrometry characterization of oxidized amyloid beta peptides as potential biomarkers of Alzheimer's disease, Rapid Commun. Mass Spectrom., 20, 911, 10.1002/rcm.2395

Brinkmalm, 2015, Explorative and targeted neuroproteomics in Alzheimer's disease, Biochim Biophys. Acta, 1854, 769, 10.1016/j.bbapap.2015.01.009

Arce-Varas, 2017, Comparison of extracellular and intracellular blood compartments highlights redox alterations in Alzheimer's and mild cognitive impairment patients, Curr. Alzheimer Res., 14, 112, 10.2174/1567205013666161010125413

Butterfield, 2006, Redox proteomics identification of oxidatively modified hippocampal proteins in mild cognitive impairment: insights into the development of Alzheimer's disease, Neurobiol. Dis., 22, 223, 10.1016/j.nbd.2005.11.002

Cheignon, 2016, Metal-catalyzed oxidation of Abeta and the resulting reorganization of Cu binding sites promote ROS production, Metallomics : Integr. Biomet. Sci., 8, 1081, 10.1039/C6MT00150E

Drew, 2017, The case for abandoning therapeutic chelation of copper ions in Alzheimer's Disease, Front Neurosci., 11, 317, 10.3389/fnins.2017.00317

Conte-Daban, 2016, How Zn can impede Cu detoxification by chelating agents in Alzheimer's disease: a proof-of-concept study, Dalton Trans., 45, 15671, 10.1039/C6DT02308H

Atrian-Blasco, 2017, Mutual interference of Cu and Zn ions in Alzheimer’s disease: perspectives at a molecular level, Dalton Trans., 46, 12750, 10.1039/C7DT01344B

Sevigny, 2016, The antibody aducanumab reduces Abeta plaques in Alzheimer's disease, Nature, 537, 50, 10.1038/nature19323