Skip to main content
Neuroscience Bulletin logoLink to Neuroscience Bulletin
. 2014 Mar 24;30(2):271–281. doi: 10.1007/s12264-013-1423-y

Oxidative stress in Alzheimer’s disease

Zhichun Chen 1,2, Chunjiu Zhong 1,2,
PMCID: PMC5562667  PMID: 24664866

Abstract

Oxidative stress plays a significant role in the pathogenesis of Alzheimer’s disease (AD), a devastating disease of the elderly. The brain is more vulnerable than other organs to oxidative stress, and most of the components of neurons (lipids, proteins, and nucleic acids) can be oxidized in AD due to mitochondrial dysfunction, increased metal levels, inflammation, and β-amyloid (Aβ) peptides. Oxidative stress participates in the development of AD by promoting Aβ deposition, tau hyperphosphorylation, and the subsequent loss of synapses and neurons. The relationship between oxidative stress and AD suggests that oxidative stress is an essential part of the pathological process, and antioxidants may be useful for AD treatment.

Keywords: Alzheimer’s disease, oxidative stress, β-amyloid, tau, metals, antioxidants

References

  • [1].Sokoloff L. Energetics of functional activation in neural tissues. Neurochem Res. 1999;24:321–329. doi: 10.1023/a:1022534709672. [DOI] [PubMed] [Google Scholar]
  • [2].Tholey G, Ledig M. Neuronal and astrocytic plasticity: metabolic aspects. Ann Med Interne (Paris) 1990;141(Suppl1):13–18. [PubMed] [Google Scholar]
  • [3].Pratico D. Oxidative stress hypothesis in Alzheimer’s disease: a reappraisal. Trends Pharmacol Sci. 2008;29:609–615. doi: 10.1016/j.tips.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • [4].Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G, Smith MA. Involvement of oxidative stress in Alzheimer disease. J Neuropathol Exp Neurol. 2006;65:631–641. doi: 10.1097/01.jnen.0000228136.58062.bf. [DOI] [PubMed] [Google Scholar]
  • [5].Pocernich C, Butterfield D. Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim Biophys Acta. 2012;1822(5):625–630. doi: 10.1016/j.bbadis.2011.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Behl C, Moosmann B. Antioxidant neuroprotection in Alzheimer’s disease as preventive and therapeutic approach. Free Radic Biol Med. 2002;33(2):182–191. doi: 10.1016/s0891-5849(02)00883-3. [DOI] [PubMed] [Google Scholar]
  • [7].Moosmann B, Behl C. Antioxidants as treatment for neurodegenerative disorders. Expert Opin Investig Drugs. 2002;11(10):1407–1435. doi: 10.1517/13543784.11.10.1407. [DOI] [PubMed] [Google Scholar]
  • [8].Torres LL, Quaglio NB, de Souza GT, Garcia RT, Dati LM, Moreira WL, et al. Peripheral oxidative stress biomarkers in mild cognitive impairment and Alzheimer’s disease. J Alzheimers Dis. 2011;26:59–68. doi: 10.3233/JAD-2011-110284. [DOI] [PubMed] [Google Scholar]
  • [9].Beal MF. Oxidative damage as an early marker of Alzheimer’s disease and mild cognitive impairment. Neurobiol Aging. 2005;26:585–586. doi: 10.1016/j.neurobiolaging.2004.09.022. [DOI] [PubMed] [Google Scholar]
  • [10].Beal M. Oxidatively modiied proteins in aging and disease. Free Radic Biol Med. 2002;32(9):797–803. doi: 10.1016/s0891-5849(02)00780-3. [DOI] [PubMed] [Google Scholar]
  • [11].Butterfield D, Kanski J. Brain protein oxidation in agerelated neurodegenerative disorders that are associated with aggregated proteins. Mech Ageing Dev. 2001;122(9):945–962. doi: 10.1016/s0047-6374(01)00249-4. [DOI] [PubMed] [Google Scholar]
  • [12].Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev. 2013;2013:316523. doi: 10.1155/2013/316523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Lovell MA, Ehmann WD, Butler SM, Markesbery WR. Elevated thiobarbituric acid-reactive substances and antioxidant enzyme activity in the brain in Alzheimer’s disease. Neurology. 1995;45:1594–1601. doi: 10.1212/wnl.45.8.1594. [DOI] [PubMed] [Google Scholar]
  • [14].Marcus DL, Thomas C, Rodriguez C, Simberkoff K, Tsai JS, Strafaci JA, et al. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer’s disease. Exp Neurol. 1998;150:40–44. doi: 10.1006/exnr.1997.6750. [DOI] [PubMed] [Google Scholar]
  • [15].Pratico D, Clark CM, Lee VM, Trojanowski JQ, Rokach J, FitzGerald GA. Increased 8,12-iso-iPF2alpha-VI in Alzheimer’s disease: correlation of a noninvasive index of lipid peroxidation with disease severity. Ann Neurol. 2000;48:809–812. [PubMed] [Google Scholar]
  • [16].Williams TI, Lynn BC, Markesbery WR, Lovell MA. Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in Mild Cognitive Impairment and early Alzheimer’s disease. Neurobiol Aging. 2006;27:1094–1099. doi: 10.1016/j.neurobiolaging.2005.06.004. [DOI] [PubMed] [Google Scholar]
  • [17].Omar RA, Chyan YJ, Andorn AC, Poeggeler B, Robakis NK, Pappolla MA. Increased expression but reduced activity of antioxidant enzymes in Alzheimer’s disease. J Alzheimers Dis. 1999;1:139–145. doi: 10.3233/jad-1999-1301. [DOI] [PubMed] [Google Scholar]
  • [18].Federico A, Cardaioli E, Da Pozzo P, Formichi P, Gallus GN, Radi E. Mitochondria, oxidative stress and neurodegeneration. J Neurol Sci. 2012;322:254–262. doi: 10.1016/j.jns.2012.05.030. [DOI] [PubMed] [Google Scholar]
  • [19].Yan MH, Wang X, Zhu X. Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med. 2013;62:90–101. doi: 10.1016/j.freeradbiomed.2012.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Ayton S, Lei P, Bush AI. Metallostasis in Alzheimer’s disease. Free Radic Biol Med. 2013;62:76–89. doi: 10.1016/j.freeradbiomed.2012.10.558. [DOI] [PubMed] [Google Scholar]
  • [21].Greenough MA, Camakaris J, Bush AI. Metal dyshomeostasis and oxidative stress in Alzheimer’s disease. Neurochem Int. 2013;62:540–555. doi: 10.1016/j.neuint.2012.08.014. [DOI] [PubMed] [Google Scholar]
  • [22].Dias-Santagata D, Fulga TA, Duttaroy A, Feany MB. Oxidative stress mediates tau-induced neurodegeneration in Drosophila. J Clin Invest. 2007;117:236–245. doi: 10.1172/JCI28769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;156:1051–1063. doi: 10.1083/jcb.200108057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Candore G, Bulati M, Caruso C, Castiglia L, Colonna-Romano G, Di Bona D, et al. Inflammation, cytokines, immune response, apolipoprotein E, cholesterol, and oxidative stress in Alzheimer disease: therapeutic implications. Rejuvenation Res. 2010;13:301–313. doi: 10.1089/rej.2009.0993. [DOI] [PubMed] [Google Scholar]
  • [25].Lee YJ, Han SB, Nam SY, Oh KW, Hong JT. Inflammation and Alzheimer’s disease. Arch Pharm Res. 2010;33:1539–1556. doi: 10.1007/s12272-010-1006-7. [DOI] [PubMed] [Google Scholar]
  • [26].Ansari MA, Scheff SW. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol. 2010;69:155–167. doi: 10.1097/NEN.0b013e3181cb5af4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Manczak M, Mao P, Calkins MJ, Cornea A, Reddy AP, Murphy MP, et al. Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer’s disease neurons. J Alzheimers Dis. 2010;20(Suppl2):S609–631. doi: 10.3233/JAD-2010-100564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature. 2004;430:631–639. doi: 10.1038/nature02621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Reddy PH. Mitochondrial oxidative damage in aging and Alzheimer’s disease: implications for mitochondrially targeted antioxidant therapeutics. J Biomed Biotechnol. 2006;2006:31372. doi: 10.1155/JBB/2006/31372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Skoumalova A, Hort J. Blood markers of oxidative stress in Alzheimer’s disease. J Cell Mol Med. 2012;16:2291–2300. doi: 10.1111/j.1582-4934.2012.01585.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Soderberg M, Edlund C, Kristensson K, Dallner G. Fatty acid composition of brain phospholipids in aging and in Alzheimer’s disease. Lipids. 1991;26:421–425. doi: 10.1007/BF02536067. [DOI] [PubMed] [Google Scholar]
  • [32].Butterfield DA, Reed T, Perluigi M, De Marco C, Coccia R, Cini C, et al. Elevated protein-bound levels of the lipid peroxidation product, 4-hydroxy-2-nonenal, in brain from persons with mild cognitive impairment. Neurosci Lett. 2006;397:170–173. doi: 10.1016/j.neulet.2005.12.017. [DOI] [PubMed] [Google Scholar]
  • [33].Keller JN, Schmitt FA, Scheff SW, Ding Q, Chen Q, Butterfield DA, et al. Evidence of increased oxidative damage in subjects with mild cognitive impairment. Neurology. 2005;64:1152–1156. doi: 10.1212/01.WNL.0000156156.13641.BA. [DOI] [PubMed] [Google Scholar]
  • [34].Roberts LJ, 2nd, Montine TJ, Markesbery WR, Tapper AR, Hardy P, Chemtob S, et al. Formation of isoprostane-like compounds (neuroprostanes) in vivo from docosahexaenoic acid. J Biol Chem. 1998;273:13605–13612. doi: 10.1074/jbc.273.22.13605. [DOI] [PubMed] [Google Scholar]
  • [35].Montine TJ, Markesbery WR, Morrow JD, Roberts LJ., 2nd Cerebrospinal fluid F2-isoprostane levels are increased in Alzheimer’s disease. Ann Neurol. 1998;44:410–413. doi: 10.1002/ana.410440322. [DOI] [PubMed] [Google Scholar]
  • [36].Singh M, Dang TN, Arseneault M, Ramassamy C. Role of by-products of lipid oxidation in Alzheimer’s disease brain: a focus on acrolein. J Alzheimers Dis. 2010;21:741–756. doi: 10.3233/JAD-2010-100405. [DOI] [PubMed] [Google Scholar]
  • [37].Ahmed N, Ahmed U, Thornalley PJ, Hager K, Fleischer G, Munch G. Protein glycation, oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer’s disease and link to cognitive impairment. J Neurochem. 2005;92:255–263. doi: 10.1111/j.1471-4159.2004.02864.x. [DOI] [PubMed] [Google Scholar]
  • [38].Butterfield DA, Reed TT, Perluigi M, De Marco C, Coccia R, Keller JN, et al. Elevated levels of 3-nitrotyrosine in brain from subjects with amnestic mild cognitive impairment: implications for the role of nitration in the progression of Alzheimer’s disease. Brain Res. 2007;1148:243–248. doi: 10.1016/j.brainres.2007.02.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol. 1994;36:747–751. doi: 10.1002/ana.410360510. [DOI] [PubMed] [Google Scholar]
  • [40].Nunomura A, Perry G, Pappolla MA, Wade R, Hirai K, Chiba S, et al. RNA oxidation is a prominent feature of vulnerable neurons in Alzheimer’s disease. J Neurosci. 1999;19:1959–1964. doi: 10.1523/JNEUROSCI.19-06-01959.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Mullaart E, Boerrigter ME, Ravid R, Swaab DF, Vijg J. Increased levels of DNA breaks in cerebral cortex of Alzheimer’s disease patients. Neurobiol Aging. 1990;11:169–173. doi: 10.1016/0197-4580(90)90542-8. [DOI] [PubMed] [Google Scholar]
  • [42].Grivennikova VG, Vinogradov AD. Generation of superoxide by the mitochondrial Complex I. Biochim Biophys Acta. 2006;1757:553–561. doi: 10.1016/j.bbabio.2006.03.013. [DOI] [PubMed] [Google Scholar]
  • [43].Tan S, Sagara Y, Liu Y, Maher P, Schubert D. The regulation of reactive oxygen species production during programmed cell death. J Cell Biol. 1998;141:1423–1432. doi: 10.1083/jcb.141.6.1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, et al. Mitochondrial abnormalities in Alzheimer’s disease. J Neurosci. 2001;21:3017–3023. doi: 10.1523/JNEUROSCI.21-09-03017.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Silva DF, Selfridge JE, Lu J, Lu E L, Cardoso SM, Swerdlow RH. Mitochondrial abnormalities in Alzheimer’s disease: possible targets for therapeutic intervention. Adv Pharmacol. 2012;64:83–126. doi: 10.1016/B978-0-12-394816-8.00003-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Mutisya EM, Bowling AC, Beal MF. Cortical cytochrome oxidase activity is reduced in Alzheimer’s disease. J Neurochem. 1994;63:2179–2184. doi: 10.1046/j.1471-4159.1994.63062179.x. [DOI] [PubMed] [Google Scholar]
  • [47].Anantharaman M, Tangpong J, Keller JN, Murphy MP, Markesbery WR, Kiningham KK, et al. Beta-amyloid mediated nitration of manganese superoxide dismutase: implication for oxidative stress in a APPNLH/NLH X PS-1P264L/P264L double knock-in mouse model of Alzheimer’s disease. Am J Pathol. 2006;168:1608–1618. doi: 10.2353/ajpath.2006.051223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Casley CS, Canevari L, Land JM, Clark JB, Sharpe MA. Beta-amyloid inhibits integrated mitochondrial respiration and key enzyme activities. J Neurochem. 2002;80:91–100. doi: 10.1046/j.0022-3042.2001.00681.x. [DOI] [PubMed] [Google Scholar]
  • [49].Wang X, Su B, Siedlak SL, Moreira PI, Fujioka H, Wang Y, et al. Amyloid-beta overproduction causes abnormal mitochondrial dynamics via differential modulation of mitochondrial fission/fusion proteins. Proc Natl Acad Sci U S A. 2008;105:19318–19323. doi: 10.1073/pnas.0804871105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Crouch PJ, Harding SM, White AR, Camakaris J, Bush AI, Masters CL. Mechanisms of A beta mediated neurodegeneration in Alzheimer’s disease. Int J Biochem Cell Biol. 2008;40:181–198. doi: 10.1016/j.biocel.2007.07.013. [DOI] [PubMed] [Google Scholar]
  • [51].Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH. 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. 2006;15:1437–1449. doi: 10.1093/hmg/ddl066. [DOI] [PubMed] [Google Scholar]
  • [52].Rodrigues CM, Sola S, Brito MA, Brondino CD, Brites D, Moura JJ. Amyloid beta-peptide disrupts mitochondrial membrane lipid and protein structure: protective role of tauroursodeoxycholate. Biochem Biophys Res Commun. 2001;281:468–474. doi: 10.1006/bbrc.2001.4370. [DOI] [PubMed] [Google Scholar]
  • [53].Apelt J, Bigl M, Wunderlich P, Schliebs R. Aging-related increase in oxidative stress correlates with developmental pattern of beta-secretase activity and beta-amyloid plaque formation in transgenic Tg2576 mice with Alzheimer-like pathology. Int J Dev Neurosci. 2004;22:475–484. doi: 10.1016/j.ijdevneu.2004.07.006. [DOI] [PubMed] [Google Scholar]
  • [54].Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH. Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet. 2006;15:1437–1449. doi: 10.1093/hmg/ddl066. [DOI] [PubMed] [Google Scholar]
  • [55].Matsuoka Y, Picciano M, La Francois J, Duff K. Fibrillar betaamyloid evokes oxidative damage in a transgenic mouse model of Alzheimer’s disease. Neuroscience. 2001;104:609–613. doi: 10.1016/s0306-4522(01)00115-4. [DOI] [PubMed] [Google Scholar]
  • [56].Takuma K, Yao J, Huang J, Xu H, Chen X, Luddy J, et al. ABAD enhances Abeta-induced cell stress via mitochondrial dysfunction. FASEB J. 2005;19:597–598. doi: 10.1096/fj.04-2582fje. [DOI] [PubMed] [Google Scholar]
  • [57].Yu T, Robotham JL, Yoon Y. Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc Natl Acad Sci U S A. 2006;103:2653–2658. doi: 10.1073/pnas.0511154103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Bonda DJ, Wang X, Perry G, Smith MA, Zhu X. Mitochondrial dynamics in Alzheimer’s disease: opportunities for future treatment strategies. Drugs Aging. 2010;27:181–192. doi: 10.2165/11532140-000000000-00000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Wang X, Su B, Fujioka H, Zhu X. Dynamin-like protein 1 reduction underlies mitochondrial morphology and distribution abnormalities in fibroblasts from sporadic Alzheimer’s disease patients. Am J Pathol. 2008;173:470–482. doi: 10.2353/ajpath.2008.071208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Baldeiras I, Santana I, Proenca MT, Garrucho MH, Pascoal R, Rodrigues A, et al. Oxidative damage and progression to Alzheimer’s disease in patients with mild cognitive impairment. J Alzheimers Dis. 2010;21:1165–1177. doi: 10.3233/jad-2010-091723. [DOI] [PubMed] [Google Scholar]
  • [61].Echtay KS. Mitochondrial uncoupling proteins—what is their physiological role? Free Radic Biol Med. 2007;43:1351–1371. doi: 10.1016/j.freeradbiomed.2007.08.011. [DOI] [PubMed] [Google Scholar]
  • [62].Wu Z, Zhang J, Zhao B. Superoxide anion regulates the mitochondrial free Ca2+ through uncoupling proteins. Antioxid Redox Signal. 2009;11:1805–1818. doi: 10.1089/ars.2009.2427. [DOI] [PubMed] [Google Scholar]
  • [63].Piccolo G, Banfi P, Azan G, Rizzuto R, Bisson R, Sandona D, et al. Biological markers of oxidative stress in mitochondrial myopathies with progressive external ophthalmoplegia. J Neurol Sci. 1991;105:57–60. doi: 10.1016/0022-510x(91)90118-q. [DOI] [PubMed] [Google Scholar]
  • [64].Smits P, Mattijssen S, Morava E, van den Brand M, Brandt F, Wijburg F, et al. Functional consequences of mitochondrial tRNA Trp and tRNA Arg mutations causing combined OXPHOS defects. Eur J Hum Genet. 2010;18:324–329. doi: 10.1038/ejhg.2009.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Wu SB, Ma YS, Wu YT, Chen YC, Wei YH. Mitochondrial DNA mutation-elicited oxidative stress, oxidative damage, and altered gene expression in cultured cells of patients with MERRF syndrome. Mol Neurobiol. 2010;41:256–266. doi: 10.1007/s12035-010-8123-7. [DOI] [PubMed] [Google Scholar]
  • [66].Ikawa M, Arakawa K, Hamano T, Nagata M, Nakamoto Y, Kuriyama M, et al. Evaluation of systemic redox states in patients carrying the MELAS A3243G mutation in mitochondrial DNA. Eur Neurol. 2012;67:232–237. doi: 10.1159/000336568. [DOI] [PubMed] [Google Scholar]
  • [67].Wong A, Cavelier L, Collins-Schramm HE, Seldin MF, McGrogan M, Savontaus M-L, et al. Differentiation-specific effects of LHON mutations introduced into neuronal NT2 cells. Hum Mol Genet. 2002;11:431–438. doi: 10.1093/hmg/11.4.431. [DOI] [PubMed] [Google Scholar]
  • [68].Deibel MA, Ehmann WD, Markesbery WR. Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer’s disease: possible relation to oxidative stress. J Neurol Sci. 1996;143:137–142. doi: 10.1016/s0022-510x(96)00203-1. [DOI] [PubMed] [Google Scholar]
  • [69].Eskici G, Axelsen PH. Copper and oxidative stress in the pathogenesis of Alzheimer’s disease. Biochemistry. 2012;51:6289–6311. doi: 10.1021/bi3006169. [DOI] [PubMed] [Google Scholar]
  • [70].Jomova K, Vondrakova D, Lawson M, Valko M. Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem. 2010;345:91–104. doi: 10.1007/s11010-010-0563-x. [DOI] [PubMed] [Google Scholar]
  • [71].Curtain CC, Ali F, Volitakis I, Cherny RA, Norton RS, Beyreuther K, et al. Alzheimer’s disease amyloid-beta binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits. J Biol Chem. 2001;276:20466–20473. doi: 10.1074/jbc.M100175200. [DOI] [PubMed] [Google Scholar]
  • [72].Opazo C, Huang X, Cherny RA, Moir RD, Roher AE, White AR, et al. Metalloenzyme-like activity of Alzheimer’s disease beta-amyloid. Cu-dependent catalytic conversion of dopamine, cholesterol, and biological reducing agents to neurotoxic H2O2. J Biol Chem. 2002;277:40302–40308. doi: 10.1074/jbc.M206428200. [DOI] [PubMed] [Google Scholar]
  • [73].Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JD, Hanson GR, et al. Cu(II) potentiation of alzheimer abeta neurotoxicity. Correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem. 1999;274:37111–37116. doi: 10.1074/jbc.274.52.37111. [DOI] [PubMed] [Google Scholar]
  • [74].Lynch T, Cherny RA, Bush AI. Oxidative processes in Alzheimer’s disease: the role of abeta-metal interactions. Exp Gerontol. 2000;35:445–451. doi: 10.1016/s0531-5565(00)00112-1. [DOI] [PubMed] [Google Scholar]
  • [75].Altamura S, Muckenthaler MU. Iron toxicity in diseases of aging: Alzheimer’s disease, Parkinson’s disease and atherosclerosis. J Alzheimers Dis. 2009;16:879–895. doi: 10.3233/JAD-2009-1010. [DOI] [PubMed] [Google Scholar]
  • [76].Pulliam JF, Jennings CD, Kryscio RJ, Davis DG, Wilson D, Montine TJ, et al. Association of HFE mutations with neurodegeneration and oxidative stress in Alzheimer’s disease and correlation with APOE. Am J Med Genet B Neuropsychiatr Genet. 2003;119B:48–53. doi: 10.1002/ajmg.b.10069. [DOI] [PubMed] [Google Scholar]
  • [77].Honda K, Casadesus G, Petersen RB, Perry G, Smith MA. Oxidative stress and redox-active iron in Alzheimer’s disease. Ann N Y Acad Sci. 2004;1012:179–182. doi: 10.1196/annals.1306.015. [DOI] [PubMed] [Google Scholar]
  • [78].Wan L, Nie G, Zhang J, Luo Y, Zhang P, Zhang Z, et al. beta-Amyloid peptide increases levels of iron content and oxidative stress in human cell and Caenorhabditis elegans models of Alzheimer disease. Free Radic Biol Med. 2011;50:122–129. doi: 10.1016/j.freeradbiomed.2010.10.707. [DOI] [PubMed] [Google Scholar]
  • [79].Sensi SL, Rapposelli IG, Frazzini V, Mascetra N. Altered oxidant-mediated intraneuronal zinc mobilization in a triple transgenic mouse model of Alzheimer’s disease. Exp Gerontol. 2008;43:488–492. doi: 10.1016/j.exger.2007.10.018. [DOI] [PubMed] [Google Scholar]
  • [80].Sensi SL, Paoletti P, Bush AI, Sekler I. Zinc in the physiology and pathology of the CNS. Nat Rev Neurosci. 2009;10:780–791. doi: 10.1038/nrn2734. [DOI] [PubMed] [Google Scholar]
  • [81].Goedert M, Spillantini MG. A century of Alzheimer’s disease. Science. 2006;314:777–781. doi: 10.1126/science.1132814. [DOI] [PubMed] [Google Scholar]
  • [82].Petersen RB, Nunomura A, Lee HG, Casadesus G, Perry G, Smith MA, et al. Signal transduction cascades associated with oxidative stress in Alzheimer’s disease. J Alzheimers Dis. 2007;11:143–152. doi: 10.3233/jad-2007-11202. [DOI] [PubMed] [Google Scholar]
  • [83].Cente M, Filipcik P, Pevalova M, Novak M. Expression of a truncated tau protein induces oxidative stress in a rodent model of tauopathy. Eur J Neurosci. 2006;24:1085–1090. doi: 10.1111/j.1460-9568.2006.04986.x. [DOI] [PubMed] [Google Scholar]
  • [84].David DC, Hauptmann S, Scherping I, Schuessel K, Keil U, Rizzu P, et al. Proteomic and functional analyses reveal a mitochondrial dysfunction in P301L tau transgenic mice. J Biol Chem. 2005;280:23802–23814. doi: 10.1074/jbc.M500356200. [DOI] [PubMed] [Google Scholar]
  • [85].Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova NN, et al. Behavioral deficit, oxidative stress, and mitochondrial dysfunction precede tau pathology in P301S transgenic mice. FASEB J. 2011;25:4063–4072. doi: 10.1096/fj.11-186650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337–351. doi: 10.1016/j.neuron.2007.01.010. [DOI] [PubMed] [Google Scholar]
  • [87].Elipenahli C, Stack C, Jainuddin S, Gerges M, Yang L, Starkov A, et al. Behavioral improvement after chronic administration of coenzyme Q10 in P301S transgenic mice. J Alzheimers Dis. 2012;28:173–182. doi: 10.3233/JAD-2011-111190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Tuppo EE, Arias HR. The role of inflammation in Alzheimer’s disease. Int J Biochem Cell Biol. 2005;37:289–305. doi: 10.1016/j.biocel.2004.07.009. [DOI] [PubMed] [Google Scholar]
  • [89].El Khoury J, Hickman SE, Thomas CA, Cao L, Silverstein SC, Loike JD. Scavenger receptor-mediated adhesion of microglia to beta-amyloid fibrils. Nature. 1996;382:716–719. doi: 10.1038/382716a0. [DOI] [PubMed] [Google Scholar]
  • [90].Johnstone M, Gearing AJ, Miller KM. A central role for astrocytes in the inflammatory response to beta-amyloid; chemokines, cytokines and reactive oxygen species are produced. J Neuroimmunol. 1999;93:182–193. doi: 10.1016/s0165-5728(98)00226-4. [DOI] [PubMed] [Google Scholar]
  • [91].Smits HA, Rijsmus A, van Loon JH, Wat JW, Verhoef J, Boven LA, et al. Amyloid-beta-induced chemokine production in primary human macrophages and astrocytes. J Neuroimmunol. 2002;127:160–168. doi: 10.1016/s0165-5728(02)00112-1. [DOI] [PubMed] [Google Scholar]
  • [92].Luo Y, Sunderland T, Roth GS, Wolozin B. Physiological levels of beta-amyloid peptide promote PC12 cell proliferation. Neurosci Lett. 1996;217:125–128. [PubMed] [Google Scholar]
  • [93].Kontush A, Schekatolina S. Vitamin E in Neurodegenerative Disorders: Alzheimer’s Disease. Ann N Y Acad Sci. 2004;1031:249–262. doi: 10.1196/annals.1331.025. [DOI] [PubMed] [Google Scholar]
  • [94].Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski A, Palmeri A, et al. Picomolar amyloid-beta positively modulates synaptic plasticity and memory in hippocampus. J Neurosci. 2008;28:14537–14545. doi: 10.1523/JNEUROSCI.2692-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60:759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
  • [96].Nunomura A, Tamaoki T, Tanaka K, Motohashi N, Nakamura M, Hayashi T, et al. Intraneuronal amyloid beta accumulation and oxidative damage to nucleic acids in Alzheimer disease. Neurobiol Dis. 2010;37:731–737. doi: 10.1016/j.nbd.2009.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Castegna A, Aksenov M, Aksenova M, Thongboonkerd V, Klein JB, Pierce WM, et al. Proteomic identification of oxidatively modified proteins in Alzheimer’s disease brain. Part I: creatine kinase BB, glutamine synthase, and ubiquitin carboxy-terminal hydrolase L-1. Free Radic Biol Med. 2002;33:562–571. doi: 10.1016/s0891-5849(02)00914-0. [DOI] [PubMed] [Google Scholar]
  • [98].Paola D, Domenicotti C, Nitti M, Vitali A, Borghi R, Cottalasso D, et al. Oxidative stress induces increase in intracellular amyloid beta-protein production and selective activation of betaI and betaII PKCs in NT2 cells. Biochem Biophys Res Commun. 2000;268:642–646. doi: 10.1006/bbrc.2000.2164. [DOI] [PubMed] [Google Scholar]
  • [99].Nunomura A, Perry G, Pappolla MA, Friedland RP, Hirai K, Chiba S, et al. Neuronal oxidative stress precedes amyloidbeta deposition in Down syndrome. J Neuropathol Exp Neurol. 2000;59:1011–1017. doi: 10.1093/jnen/59.11.1011. [DOI] [PubMed] [Google Scholar]
  • [100].Oda A, Tamaoka A, Araki W. Oxidative stress up-regulates presenilin 1 in lipid rafts in neuronal cells. J Neurosci Res. 2010;88:1137–1145. doi: 10.1002/jnr.22271. [DOI] [PubMed] [Google Scholar]
  • [101].Tamagno E, Bardini P, Obbili A, Vitali A, Borghi R, Zaccheo D, et al. Oxidative stress increases expression and activity of BACE in NT2 neurons. Neurobiol Dis. 2002;10:279–288. doi: 10.1006/nbdi.2002.0515. [DOI] [PubMed] [Google Scholar]
  • [102].Yoo MH, Gu X, Xu XM, Kim JY, Carlson BA, Patterson AD, et al. Delineating the role of glutathione peroxidase 4 in protecting cells against lipid hydroperoxide damage and in Alzheimer’s disease. Antioxid Redox Signal. 2010;12:819–827. doi: 10.1089/ars.2009.2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Tamagno E, Guglielmotto M, Aragno M, Borghi R, Autelli R, Giliberto L, et al. Oxidative stress activates a positive feedback between the γ- and β-secretase cleavages of the β-amyloid precursor protein. J Neurochem. 2008;104:683–695. doi: 10.1111/j.1471-4159.2007.05072.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Cole GM, Teter B, Frautschy SA. Neuroprotective effects of curcumin. Adv Exp Med Biol. 2007;595:197–212. doi: 10.1007/978-0-387-46401-5_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Mandel SA, Amit T, Kalfon L, Reznichenko L, Youdim MB. Targeting multiple neurodegenerative diseases etiologies with multimodal-acting green tea catechins. J Nutr. 2008;138:1578S–1583S. doi: 10.1093/jn/138.8.1578S. [DOI] [PubMed] [Google Scholar]
  • [106].Smith JV, Luo Y. Studies on molecular mechanisms of Ginkgo biloba extract. Appl Microbiol Biotechnol. 2004;64:465–472. doi: 10.1007/s00253-003-1527-9. [DOI] [PubMed] [Google Scholar]
  • [107].Dumont M, Wille E, Stack C, Calingasan NY, Beal MF, Lin MT. Reduction of oxidative stress, amyloid deposition, and memory deficit by manganese superoxide dismutase overexpression in a transgenic mouse model of Alzheimer’s disease. FASEB J. 2009;23:2459–2466. doi: 10.1096/fj.09-132928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Murakami K, Murata N, Noda Y, Tahara S, Kaneko T, Kinoshita N, et al. SOD1 (copper/zinc superoxide dismutase) deficiency drives amyloid beta protein oligomerization and memory loss in mouse model of Alzheimer disease. J Biol Chem. 2011;286:44557–44568. doi: 10.1074/jbc.M111.279208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Gamblin TC, King ME, Kuret J, Berry RW, Binder LI. Oxidative regulation of fatty acid-induced tau polymerization. Biochemistry. 2000;39:14203–14210. doi: 10.1021/bi001876l. [DOI] [PubMed] [Google Scholar]
  • [110].Schweers O, Mandelkow EM, Biernat J, Mandelkow E. Oxidation of cysteine-322 in the repeat domain of microtubule-associated protein tau controls the in vitro assembly of paired helical filaments. Proc Natl Acad Sci U S A. 1995;92:8463–8467. doi: 10.1073/pnas.92.18.8463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Melov S, Adlard PA, Morten K, Johnson F, Golden TR, Hinerfeld D, et al. Mitochondrial oxidative stress causes hyperphosphorylation of tau. PLoS One. 2007;2:e536. doi: 10.1371/journal.pone.0000536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Goedert M, Hasegawa M, Jakes R, Lawler S, Cuenda A, Cohen P. Phosphorylation of microtubule-associated protein tau by stress-activated protein kinases. FEBS Lett. 1997;409:57–62. doi: 10.1016/s0014-5793(97)00483-3. [DOI] [PubMed] [Google Scholar]
  • [113].Wadsworth TL, Bishop JA, Pappu AS, Woltjer RL, Quinn JF. Evaluation of coenzyme Q as an antioxidant strategy for Alzheimer’s disease. J Alzheimer Dis. 2008;14:225–234. doi: 10.3233/jad-2008-14210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Yang X, Dai G, Li G, Yang E. Coenzyme Q10 reduces β-amyloid plaque in an APP/PS1 transgenic mouse model of Alzheimer’s disease. J Mol Neurosci. 2010;41:110–113. doi: 10.1007/s12031-009-9297-1. [DOI] [PubMed] [Google Scholar]
  • [115].Yang X, Yang Y, Li G, Wang J, Yang E. Coenzyme Q10 attenuates β-amyloid pathology in the aged transgenic mice with Alzheimer Presenilin 1 mutation. J Mol Neurosci. 2008;34:165–171. doi: 10.1007/s12031-007-9033-7. [DOI] [PubMed] [Google Scholar]
  • [116].Gutzmann H, Kuhl KP, Hadler D, Rapp MA. Safety and efficacy of idebenone versus tacrine in patients with Alzheimer’s disease: results of a randomized, double-blind, parallel-group multicenter study. Pharmacopsychiatry. 2002;35:12–18. doi: 10.1055/s-2002-19833. [DOI] [PubMed] [Google Scholar]
  • [117].Senin U, Parnetti L, Barbagallo-Sangiorgi G, Bartorelli L, Bocola V, Capurso A, et al. Idebenone in senile dementia of Alzheimer type: a multicentre study. Arch Gerontol Geriatr. 1992;15:249–260. doi: 10.1016/0167-4943(92)90060-h. [DOI] [PubMed] [Google Scholar]
  • [118].Thal LJ, Grundman M, Berg J, Ernstrom K, Margolin R, Pfeiffer E, et al. Idebenone treatment fails to slow cognitive decline in Alzheimer’s disease. Neurology. 2003;61:1498–1502. doi: 10.1212/01.wnl.0000096376.03678.c1. [DOI] [PubMed] [Google Scholar]
  • [119].Smith RAJ, Murphy MP. Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci. 2010;1201:96–103. doi: 10.1111/j.1749-6632.2010.05627.x. [DOI] [PubMed] [Google Scholar]
  • [120].Okun I, Tkachenko SE, Khvat A, Mitkin O, Kazey V, Ivachtchenko AV. From anti-allergic to anti-Alzheimer’s: molecular pharmacology of Dimebon. Curr Alzheimer Res. 2010;7:97–112. doi: 10.2174/156720510790691100. [DOI] [PubMed] [Google Scholar]
  • [121].Doody RS, Gavrilova SI, Sano M, Thomas RG, Aisen PS, Bachurin SO, et al. Effect of dimebon on cognition, activities of daily living, behaviour, and global function in patients with mild-to-moderate Alzheimer’s disease: a randomised, doubleblind, placebo-controlled study. Lancet. 2008;372:207–215. doi: 10.1016/S0140-6736(08)61074-0. [DOI] [PubMed] [Google Scholar]
  • [122].Jones RW. Dimebon disappointment. Alzheimers Res Ther. 2010;2:25. doi: 10.1186/alzrt49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Baum L, Lam CW, Cheung SK, Kwok T, Lui V, Tsoh J, et al. Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol. 2008;28:110–113. doi: 10.1097/jcp.0b013e318160862c. [DOI] [PubMed] [Google Scholar]
  • [124].Pettegrew JW, Klunk WE, Panchalingam K, Kanfer JN, McClure RJ. Clinical and neurochemical effects of acetyl-Lcarnitine in Alzheimer’s disease. Neurobiol Aging. 1995;16:1–4. doi: 10.1016/0197-4580(95)80001-8. [DOI] [PubMed] [Google Scholar]
  • [125].Thal LJ, Carta A, Clarke WR, Ferris SH, Friedland RP, Petersen RC, et al. A 1-year multicenter placebo-controlled study of acetyl-L-carnitine in patients with Alzheimer’s disease. Neurology. 1996;47:705–711. doi: 10.1212/wnl.47.3.705. [DOI] [PubMed] [Google Scholar]
  • [126].Thal LJ, Calvani M, Amato A, Carta A, Group ftA-l-CS A 1-year controlled trial of acetyl-l-carnitine in early-onset AD. Neurology. 2000;55:805–810. doi: 10.1212/wnl.55.6.805. [DOI] [PubMed] [Google Scholar]
  • [127].Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grundman M, et al. A Controlled Trial of selegiline, alphatocopherol, or both as treatment for Alzheimer’s disease. N Engl J. 1997;336:1216–1222. doi: 10.1056/NEJM199704243361704. [DOI] [PubMed] [Google Scholar]
  • [128].Abramova NA, Cassarino DS, Khan SM, Painter TW, Bennett JP. Inhibition by R(+) or S(−) pramipexole of caspase activation and cell death induced by methylpyridinium ion or beta amyloid peptide in SH-SY5Y neuroblastoma. J Neurosci Res. 2002;67:494–500. doi: 10.1002/jnr.10127. [DOI] [PubMed] [Google Scholar]
  • [129].Manczak M, Mao P, Calkins MJ, Cornea A, Reddy AP, Murphy MP, et al. Mitochondria-targeted antioxidants protect against amyloid-β toxicity in Alzheimer’s disease neurons. J Alzheimer Dis. 2010;20:609–631. doi: 10.3233/JAD-2010-100564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Chen Z, Zhong C. Decoding Alzheimer’s disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol. 2013;108:21–43. doi: 10.1016/j.pneurobio.2013.06.004. [DOI] [PubMed] [Google Scholar]

Articles from Neuroscience Bulletin are provided here courtesy of Springer

RESOURCES