Why pleiotropic interventions are needed for Alzheimer's disease

Sally A Frautschy, Greg M Cole, Sally A Frautschy, Greg M Cole

Abstract

Alzheimer's disease (AD) involves a complex pathological cascade thought to be initially triggered by the accumulation of beta-amyloid (Abeta) peptide aggregates or aberrant amyloid precursor protein (APP) processing. Much is known of the factors initiating the disease process decades prior to the onset of cognitive deficits, but an unclear understanding of events immediately preceding and precipitating cognitive decline is a major factor limiting the rapid development of adequate prevention and treatment strategies. Multiple pathways are known to contribute to cognitive deficits by disruption of neuronal signal transduction pathways involved in memory. These pathways are altered by aberrant signaling, inflammation, oxidative damage, tau pathology, neuron loss, and synapse loss. We need to develop stage-specific interventions that not only block causal events in pathogenesis (aberrant tau phosphorylation, Abeta production and accumulation, and oxidative damage), but also address damage from these pathways that will not be reversed by targeting prodromal pathways. This approach would not only focus on blocking early events in pathogenesis, but also adequately correct for loss of synapses, substrates for neuroprotective pathways (e.g., docosahexaenoic acid), defects in energy metabolism, and adverse consequences of inappropriate compensatory responses (aberrant sprouting). Monotherapy targeting early single steps in this complicated cascade may explain disappointments in trials with agents inhibiting production, clearance, or aggregation of the initiating Abeta peptide or its aggregates. Both plaque and tangle pathogenesis have already reached AD levels in the more vulnerable brain regions during the "prodromal" period prior to conversion to "mild cognitive impairment (MCI)." Furthermore, many of the pathological events are no longer proceeding in series, but are going on in parallel. By the MCI stage, we stand a greater chance of success by considering pleiotropic drugs or cocktails that can independently limit the parallel steps of the AD cascade at all stages, but that do not completely inhibit the constitutive normal functions of these pathways. Based on this hypothesis, efforts in our laboratories have focused on the pleiotropic activities of omega-3 fatty acids and the anti-inflammatory, antioxidant, and anti-amyloid activity of curcumin in multiple models that cover many steps of the AD pathogenic cascade (Cole and Frautschy, Alzheimers Dement 2:284-286, 2006).

Figures

Fig. 1
Fig. 1
Stage-dependent pathogenesis: This figure illustrates the complexity of different pathogenic events (inflammation, synaptic loss, oxidative damage in addition to imaging, and CSF biomarkers) at different stages, showing that there is extensive tau and Aβ pathology prior to symptoms. Identifying plasma or CSF biomarkers specific for targeted pathway (inflammation, neurodegeneration) is important for the rapid development of treatments and early interventions
Fig. 2
Fig. 2
Hypothetical Alzheimer cascade: Argument for pleiotropic targeting. This schematic diagram illustrates a refinement of the amyloid cascade, suggesting that more than one pathogenic pathway contributes to memory loss and demonstrating how anti-Aβ or other monotherapies would not succeed on their own. Examples of such pathogenic pathways that need to be targeted are oxidative damage (which depletes membrane n-3 fatty acids like DHA), aberrant inflammation, and pTau accumulation. In agreement with the “baptists,” Aβ accumulation is a triggering event. But in agreement with the “taoists,” both tau aggregates and increased pTau are likely to be major contributors to memory deficits (even though tangle pathology is not specific to AD). Nevertheless, reducing tau aggregation is unlikely to be sufficient since other events including impaired energy metabolism and aberrant are contributing to memory deficits. Compounds like curcumin or DHA or both are likely to be effective in targeting multiple steps

References

    1. Cole GM, Frautschy SA. Commentary on “Cytoskeletal modulators and pleiotropic strategies for Alzheimer drug discovery.” Pleiotropic approaches to Alzheimer's and other diseases of aging. Alzheimers Dement. 2006;2:284–286.
    1. Katzman R, Terry R, DeTeresa R, Brown T, Davies P, Fuld P, Renbing X, Peck A. Clinical, pathological and neurochemical changes in dementia: a subgroup with preserved mental status and numerous neocortical plaques. Ann Neurol. 1988;23:138–144.
    1. Price JL, Morris JC. Tangles and plaques in nondemented aging and “preclinical” Alzheimer's disease. Ann Neurol. 1999;45:358–368.
    1. Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Ramsey K, Caselli RJ, Kukull WA, McKeel D, Morris JC, Hulette CM, Schmechel D, Reiman EM, Rogers J, Stephan DA. Neuronal gene expression in non-demented individuals with intermediate Alzheimer's Disease neuropathology. Neurobiol Aging. 2008;31:549–566.
    1. Jack CR, Jr, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ. Hypothetical model of dynamic biomarkers of the Alzheimer's pathological cascade. Lancet Neurol. 2010;9:119–128.
    1. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA, Katzman R. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–580.
    1. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356.
    1. Morris JC, Storandt M, McKeel DW, Jr, Rubin EH, Price JL, Grant EA, Berg L. Cerebral amyloid deposition and diffuse plaques in “normal” aging: evidence for presymptomatic and very mild Alzheimer's disease. Neurology. 1996;46:707–719.
    1. Hardy JA, Higgins GA. Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992;256:184–185.
    1. Castellani RJ, Lee HG, Siedlak SL, Nunomura A, Hayashi T, Nakamura M, Zhu X, Perry G, Smith MA. Reexamining Alzheimer's disease: evidence for a protective role for amyloid-β protein precursor and amyloid-β. J Alzheimers Dis. 2009;18:447–452.
    1. Lee HG, Casadesus G, Zhu X, Takeda A, Perry G, Smith MA. Challenging the amyloid cascade hypothesis: senile plaques and amyloid-β as protective adaptations to Alzheimer disease. Ann N Y Acad Sci. 2004;1019:1–4.
    1. Ishida A, Furukawa K, Jeffrey NK, Mattson MP. Secreted form of b-amyloid precursor protein shifts the frequency dependency for induction of LTD, and enhances LTP in hippocampal slices. NeuroReport. 1997;8:2133–2137.
    1. Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski A, Palmeri A, Arancio O. Picomolar amyloid-β positively modulates synaptic plasticity and memory in hippocampus. J Neurosci. 2008;28:14537–14545.
    1. Cole GM, Frautschy SA. Church baptizes Joseph and Perry. Eccentric views absolved. Neurobiol Aging. 2001;22:147–150.
    1. Pimplikar SW. Reassessing the amyloid cascade hypothesis of Alzheimer's disease. Int J Biochem Cell Biol. 2009;41:1261–1268.
    1. Morihara T, Chu T, Ubeda O, Beech W, Cole GM. Selective inhibition of Aβ42 production by NSAID R-enantiomers. J Neurochem. 2002;83:1–4.
    1. Wilcock GK, Black SE, Hendrix SB, Zavitz KH, Swabb EA, Laughlin MA. Efficacy and safety of tarenflurbil in mild to moderate Alzheimer's disease: a randomised phase II trial. Lancet Neurol. 2008;7:483–493.
    1. Green RC, Schneider LS, Amato DA, Beelen AP, Wilcock G, Swabb EA, Zavitz KH. Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA. 2009;302:2557–2564.
    1. Schwab C, Hosokawa M, McGeer PL. Transgenic mice overexpressing amyloid β protein are an incomplete model of Alzheimer disease. Exp Neurol. 2004;188:52–64.
    1. Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CC, Yang A, Gallagher M, Ashe KH. A specific amyloid-β assembly in the brain impairs memory. Nature. 2006;440:352–357.
    1. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539.
    1. Murray CA, Lynch MA. Evidence that increased hippocampal expression of the cytokine interleukin-1 β is a common trigger for age- and stress-induced impairments in long-term potentiation. J Neurosci. 1998;18:2974–2981.
    1. Hauss-Wegrzyniak B, Vannucchi MG, Wenk GL. Behavioral and ultrastructural changes induced by chronic neuroinflammation in young rats. Brain Res. 2000;859:157–166.
    1. Hutton M, Lewis J, Dickson D, Yen SH, McGowan E. Analysis of tauopathies with transgenic mice. Trends Mol Med. 2001;7:467–470.
    1. Ramsden M, Kotilinek L, Forster C, Paulson J, McGowan E, SantaCruz K, Guimaraes A, Yue M, Lewis J, Carlson G, Hutton M, Ashe KH. Age-dependent neurofibrillary tangle formation, neuron loss, and memory impairment in a mouse model of human tauopathy (P301L) J Neurosci. 2005;25:10637–10647.
    1. Kimura T, Yamashita S, Fukuda T, Park JM, Murayama M, Mizoroki T, Yoshiike Y, Sahara N, Takashima A. Hyperphosphorylated tau in parahippocampal cortex impairs place learning in aged mice expressing wild-type human tau. EMBO J. 2007;26:5143–5152.
    1. Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, Jones RW, Bullock R, Love S, Neal JW, Zotova E, Nicoll JA. Long-term effects of Aβ42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372:216–223.
    1. Kokjohn TA, Roher AE. Antibody responses, amyloid-β peptide remnants and clinical effects of AN-1792 immunization in patients with AD in an interrupted trial. CNS Neurol Disord Drug Targets. 2009;8:88–97.
    1. St George-Hyslop PH, Morris JC. Will anti-amyloid therapies work for Alzheimer's disease? Lancet. 2008;372:180–182.
    1. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol (Berl) 1991;82:239–259.
    1. Cummings JL, Cole G. Alzheimer disease. JAMA. 2002;287:2335–2338.
    1. Khlistunova I, Biernat J, Wang Y, Pickhardt M, von Bergen M, Gazova Z, Mandelkow E, Mandelkow EM. Inducible expression of Tau repeat domain in cell models of tauopathy: aggregation is toxic to cells but can be reversed by inhibitor drugs. J Biol Chem. 2006;281:1205–1214.
    1. Ballatore C, Lee VM, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer's disease and related disorders. Nat Rev Neurosci. 2007;8:663–672.
    1. Iqbal K, Liu F, Gong CX, Alonso AD, Grundke-Iqbal I. Mechanisms of tau-induced neurodegeneration. Acta Neuropathol. 2009;118:53–69.
    1. Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E. Development of tau aggregation inhibitors for Alzheimer's disease. Angew Chem Int Ed Engl. 2009;48:1740–1752.
    1. Zhang B, Maiti A, Shively S, Lakhani F, McDonald-Jones G, Bruce J, Lee EB, Xie SX, Joyce S, Li C, Toleikis PM, Lee VM, Trojanowski JQ. Microtubule-binding drugs offset tau sequestration by stabilizing microtubules and reversing fast axonal transport deficits in a tauopathy model. Proc Natl Acad Sci USA. 2005;102:227–231.
    1. Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29:95–102.
    1. Hu S, Begum AN, Jones MR, Oh MS, Beech WK, Beech BH, Yang F, Chen P, Ubeda OJ, Kim PC, Davies P, Ma Q, Cole GM, Frautschy SA. GSK3 inhibitors show benefits in an Alzheimer's disease (AD) model of neurodegeneration but adverse effects in control animals. Neurobiol Dis. 2009;33:193–206.
    1. Smith MA, Perry G, Richey PL, Sayre LM, Anderson VE, Beal MF, Kowall N. Oxidative damage in Alzheimer's. Nature. 1996;382:120–121.
    1. Reich EE, Markesbery WR, Roberts LJ, 2nd, Swift LL, Morrow JD, Montine TJ. Brain regional quantification of F-ring and D-/E-ring isoprostanes and neuroprostanes in Alzheimer's disease. Am J Pathol. 2001;158:293–297.
    1. Montine KS, Kim PJ, Olson SJ, Markesbery WR, Montine TJ. 4-Hydroxy-2-nonenal pyrrole adducts in human neurodegenerative disease. J Neuropathol Exp Neurol. 1997;56:866–871.
    1. Quinn JF, Montine KS, Moore M, Morrow JD, Kaye JA, Montine TJ. Suppression of longitudinal increase in CSF F2-isoprostanes in Alzheimer's disease. J Alzheimers Dis. 2004;6:93–97.
    1. Behl C, Davis JB, Lesley R, Schubert D. Hydrogen peroxide mediates amyloid β-protein toxicity. Cell. 1994;77:817–827.
    1. Behl C, Davis J, Cole GM, Schubert D. Vitamin E protects nerve cells from amyloid β-protein toxicity. Biochem Biophys Res Commun. 1992;186:944–950.
    1. Sung S, Yao Y, Uryu K, Yang H, Lee VM, Trojanowski JQ, Pratico D. Early vitamin E supplementation in young but not aged mice reduces Aβ levels and amyloid deposition in a transgenic model of Alzheimer's disease. FASEB J. 2004;18:323–325.
    1. Siedlak SL, Casadesus G, Webber KM, Pappolla MA, Atwood CS, Smith MA, Perry G. Chronic antioxidant therapy reduces oxidative stress in a mouse model of Alzheimer's disease. Free Radic Res. 2009;43:156–164.
    1. Shinto L, Quinn J, Montine T, Baldauf-Wagner S, Oken B, Bourdette D, Kaye J. Omega-3 fatty acids and lipoic acid in Alzheimer's disease. Neurology. 2008;70:A393.
    1. Zhu X, Castellani RJ, Takeda A, Nunomura A, Atwood CS, Perry G, Smith MA. Differential activation of neuronal ERK, JNK/SAPK and p38 in Alzheimer disease: the ‘two hit’ hypothesis. Mech Ageing Dev. 2001;123:39–46.
    1. Liang X, Wang Q, Hand T, Wu L, Breyer RM, Montine TJ, Andreasson K. Deletion of the prostaglandin E2 EP2 receptor reduces oxidative damage and amyloid burden in a model of Alzheimer's disease. J Neurosci. 2005;25:10180–10187.
    1. Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Aggarwal N, Wilson RS, Wilson RS, Scherr PA. Dietary intake of antioxidant nutrients and the risk of incident Alzheimer disease in a biracial community study. JAMA. 2002;287:33230–33237.
    1. Zandi PP, Anthony JC, Khachaturian AS, Stone SV, Gustafson D, Tschanz JT, Norton MC, Welsh-Bohmer KA, Breitner JC. Reduced risk of Alzheimer disease in users of antioxidant vitamin supplements: the Cache County Study. Arch Neurol. 2004;61:82–88.
    1. Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grundman M, Woodbury P, Growdon J, Cotman CW, Pfeiffer E, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. N Engl J Med. 1997;336:1216–1222.
    1. Lu PH, Edland SD, Teng E, Tingus K, Petersen RC, Cummings JL. Donepezil delays progression to AD in MCI subjects with depressive symptoms. Neurology. 2009;72:2115–2121.
    1. Wolf G. Estimation of the human daily requirement of vitamin E by turnover kinetics of labeled RRR-alpha-tocopherol. Nutr Rev. 2007;65:46–48.
    1. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NE, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T. Inflammation and Alzheimer's disease. Neurobiol Aging. 2000;21:383–421.
    1. Cole GM, Morihara T, Lim GP, Yang F, Begum A, Frautschy SA. NSAID and antioxidant prevention of Alzheimer's disease: lessons from in vitro and animal models. Ann N Y Acad Sci. 2004;1035:68–84.
    1. Morihara T, Teter B, Yang F, Lim GP, Boudinot S, Boudinot FD, Frautschy SA, Cole GM. Ibuprofen suppresses interleukin-1β induction of pro-amyloidogenic alpha1-antichymotrypsin to ameliorate β-amyloid (Aβ) pathology in Alzheimer's models. Neuropsychopharmacology. 2005;30:1111–1120.
    1. Sastre M, Dewachter I, Landreth GE, Willson TM, Klockgether T, van Leuven F, Heneka MT. Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of β-secretase. J Neurosci. 2003;23:9796–9804.
    1. Heneka MT, Sastre M, Dumitrescu-Ozimek L, Hanke A, Dewachter I, Kuiperi C, O'Banion K, Klockgether T, Van Leuven F, Landreth GE. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Aβ1-42 levels in APPV717I transgenic mice. Brain. 2005;128:1442–1453.
    1. Sastre M, Dewachter I, Rossner S, Bogdanovic N, Rosen E, Borghgraef P, Evert BO, Dumitrescu-Ozimek L, Thal DR, Landreth G, Walter J, Klockgether T, van Leuven F, Heneka MT. Nonsteroidal anti-inflammatory drugs repress β-secretase gene promoter activity by the activation of PPARgamma. Proc Natl Acad Sci USA. 2006;103:443–448.
    1. Moore AH, Wu M, Shaftel SS, Graham KA, O'Banion MK. Sustained expression of interleukin-1β in mouse hippocampus impairs spatial memory. Neuroscience. 2009;164:1484–1495.
    1. Barrientos RM, Frank MG, Hein AM, Higgins EA, Watkins LR, Rudy JW, Maier SF. Time course of hippocampal IL-1 β and memory consolidation impairments in aging rats following peripheral infection. Brain Behav Immun. 2009;23:46–54.
    1. Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23:1605–1611.
    1. Webster SD, Yang AJ, Margol L, Garzon-Rodriguez W, Glabe CG, Tenner AJ. Complement component C1q modulates the phagocytosis of Aβ by microglia. Exp Neurol. 2000;161:127–138.
    1. Wyss-Coray T, Yan F, Lin AH, Lambris JD, Alexander JJ, Quigg RJ, Masliah E. Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer's mice. Proc Natl Acad Sci USA. 2002;99:10837–10842.
    1. Shaftel SS, Kyrkanides S, Olschowka JA, Miller JN, Johnson RE, O'Banion MK. Sustained hippocampal IL-1 β overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007;117:1595–1604.
    1. Mrak RE, Griffin WS. Glia and their cytokines in progression of neurodegeneration. Neurobiol Aging. 2005;26:349–354.
    1. Frautschy SA, Yang F, Irizarry M, Hyman B, Saido TC, Hsiao K, Cole GM. Microglial response to amyloid plaques in APPsw transgenic mice. Am J Pathol. 1998;152:307–317.
    1. Frautschy S, Cole GM, Baird A. Phagocytosis and deposition of vascular β-amyloid in rat brains injected with Alzheimer β-amyloid. Am J Pathol. 1992;140:1389–1399.
    1. Wisniewski T, Konietzko U. Amyloid-β immunisation for Alzheimer's disease. Lancet Neurol. 2008;7:805–811.
    1. Head E, Pop V, Vasilevko V, Hill M, Saing T, Sarsoza F, Nistor M, Christie LA, Milton S, Glabe C, Barrett E, Cribbs D. A two-year study with fibrillar β-amyloid (Aβ) immunization in aged canines: effects on cognitive function and brain Aβ. J Neurosci. 2008;28:3555–3566.
    1. McGeer EG, McGeer PL. Neuroinflammation in Alzheimer's disease and mild cognitive impairment: a field in its infancy. J Alzheimers Dis. 2010;19:355–361.
    1. Webster S, Lue LF, Brachova L, Tenner AJ, McGeer PL, Terai K, Walker DG, Bradt B, Cooper NR, Rogers J. Molecular and cellular characterization of the membrane attack complex, C5b-9, in Alzheimer's disease. Neurobiol Aging. 1997;18:415–421.
    1. Webster S, Bradt B, Rogers J, Cooper N. Aggregation state-dependent activation of the classical complement pathway by the amyloid b peptide. J Neurochem. 1997;69:388–398.
    1. Mrak RE, Griffin WS. Potential inflammatory biomarkers in Alzheimer's disease. J Alzheimers Dis. 2005;8:369–375.
    1. Sapolsky RM. Glucocorticoid toxicity in the hippocampus: reversal by supplementation with brain fuels. J Neurosci. 1986;6:2240–2244.
    1. Qi D, Rodrigues B. Glucocorticoids produce whole body insulin resistance with changes in cardiac metabolism. Am J Physiol Endocrinol Metab. 2007;292:E654–E667.
    1. Craft S. The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol. 2009;66:300–305.
    1. Lim GP, Yang F, Chu T, Gahtan E, Ubeda O, Beech W, Overmier JB, Hsiao Ashe K, Frautschy SA, Cole GM. Ibuprofen effects on Alzheimer pathology and open field activity in APPsw transgenic mice. Neurobiol Aging. 2001;22:983–991.
    1. McGeer PL, McGeer EG. NSAIDs and Alzheimer disease: epidemiological, animal model and clinical studies. Neurobiol Aging. 2007;28:639–647.
    1. Vlad SC, Miller DR, Kowall NW, Felson DT. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;70:1672–1677.
    1. Aisen PS. The inflammatory hypothesis of Alzheimer disease: dead or alive? Alzheimer Dis Assoc Disord. 2008;22:4–5.
    1. Martin BK, Szekely C, Brandt J, Piantadosi S, Breitner JC, Craft S, Evans D, Green R, Mullan M. Cognitive function over time in the Alzheimer's Disease Anti-inflammatory Prevention Trial (ADAPT): results of a randomized, controlled trial of naproxen and celecoxib. Arch Neurol. 2008;65:896–905.
    1. Laino C. In follow-up analysis of clinical trial, NSAIDs seem to preserve cognitive function in patients with healthy brains. Neurol Today. 2009;9:21–22.
    1. Szekely CA, Thorne JE, Zandi PP, Ek M, Messias E, Breitner JC, Goodman SN. Nonsteroidal anti-inflammatory drugs for the prevention of Alzheimer's disease: a systematic review. Neuroepidemiology. 2004;23:159–169.
    1. Lim GP, Yang F, Chu T, Chen P, Beech W, Teter B, Tran T, Ubeda O, Ashe KH, Frautschy SA, Cole GM. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer's disease. J Neurosci. 2000;20(15):5709–5714.
    1. Yan Q, Zhang J, Liu H, Babu-Khan S, Vassar R, Biere AL, Citron M, Landreth G. Anti-inflammatory drug therapy alters β-amyloid processing and deposition in an animal model of Alzheimer's disease. J Neurosci. 2003;23:7504–7509.
    1. McKee AC, Carreras I, Hossain L, Ryu H, Klein WL, Oddo S, LaFerla FM, Jenkins BG, Kowall NW, Dedeoglu A. Ibuprofen reduces Aβ, hyperphosphorylated tau and memory deficits in Alzheimer mice. Brain Res. 2008;1207:225–236.
    1. Pasqualetti P, Bonomini C, Dal Forno G, Paulon L, Sinforiani E, Marra C, Zanetti O, Rossini PM. A randomized controlled study on effects of ibuprofen on cognitive progression of Alzheimer's disease. Aging Clin Exp Res. 2009;21:102–110.
    1. Hoozemans JJ, Rozemuller JM, van Haastert ES, Veerhuis R, Eikelenboom P. Cyclooxygenase-1 and -2 in the different stages of Alzheimer's disease pathology. Curr Pharm Des. 2008;14:1419–1427.
    1. Sanchez-Mejia RO, Newman JW, Toh S, Yu GQ, Zhou Y, Halabisky B, Cisse M, Scearce-Levie K, Cheng IH, Gan L, Palop JJ, Bonventre JV, Mucke L. Phospholipase A2 reduction ameliorates cognitive deficits in a mouse model of Alzheimer's disease. Nat Neurosci. 2008;11:1311–1318.
    1. Shelat PB, Chalimoniuk M, Wang JH, Strosznajder JB, Lee JC, Sun AY, Simonyi A, Sun GY. Amyloid β peptide and NMDA induce ROS from NADPH oxidase and AA release from cytosolic phospholipase A2 in cortical neurons. J Neurochem. 2008;106:45–55.
    1. Lin TN, Wang Q, Simonyi A, Chen JJ, Cheung WM, He YY, Xu J, Sun AY, Hsu CY, Sun GY. Induction of secretory phospholipase A2 in reactive astrocytes in response to transient focal cerebral ischemia in the rat brain. J Neurochem. 2004;90:637–645.
    1. Jensen MD, Sheng W, Simonyi A, Johnson GS, Sun AY, Sun GY. Involvement of oxidative pathways in cytokine-induced secretory phospholipase A2-IIA in astrocytes. Neurochem Int. 2009;55:362–368.
    1. Clerici F, Del Sole A, Chiti A, Maggiore L, Lecchi M, Pomati S, Mosconi L, Lucignani G, Mariani C. Differences in hippocampal metabolism between amnestic and non-amnestic MCI subjects: automated FDG-PET image analysis. Q J Nucl Med Mol Imaging. 2009;53:646–657.
    1. Mosconi L, Perani D, Sorbi S, Herholz K, Nacmias B, Holthoff V, Salmon E, Baron JC, De Cristofaro MT, Padovani A, Borroni B, Franceschi M, Bracco L, Pupi A. MCI conversion to dementia and the APOE genotype: a prediction study with FDG-PET. Neurology. 2004;63:2332–2340.
    1. Haier RJ, Alkire MT, White NS, Uncapher MR, Head E, Lott IT, Cotman CW. Temporal cortex hypermetabolism in Down syndrome prior to the onset of dementia. Neurology. 2003;61:1673–1679.
    1. Becker JT, Mintun MA, Aleva K, Wiseman MB, Nichols T, Dekosky ST. Alterations in functional neuroanatomical connectivity in Alzheimer's disease. Positron emission tomography of auditory verbal short-term memory. Ann N Y Acad Sci. 1996;777:239–242.
    1. Becker JT, Mintun MA, Aleva K, Wiseman MB, Nichols T, DeKosky ST. Compensatory reallocation of brain resources supporting verbal episodic memory in Alzheimer's disease. Neurology. 1996;46:692–700.
    1. Backman L, Andersson JL, Nyberg L, Winblad B, Nordberg A, Almkvist O. Brain regions associated with episodic retrieval in normal aging and Alzheimer's disease. Neurology. 1999;52:1861–1870.
    1. Cohen AD, Price JC, Weissfeld LA, James J, Rosario BL, Bi W, Nebes RD, Saxton JA, Snitz BE, Aizenstein HA, Wolk DA, Dekosky ST, Mathis CA, Klunk WE. Basal cerebral metabolism may modulate the cognitive effects of Aβ in mild cognitive impairment: an example of brain reserve. J Neurosci. 2009;29:14770–14778.
    1. Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM. Amyloid-β dynamics are regulated by orexin and the sleep–wake cycle. Science. 2009;326:1005–1007.
    1. Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD. Mitochondrial bioenergetic deficit precedes Alzheimer's pathology in female mouse model of Alzheimer's disease. Proc Natl Acad Sci USA. 2009;106:14670–14675.
    1. Bigl M, Bruckner MK, Arendt T, Bigl V, Eschrich K. Activities of key glycolytic enzymes in the brains of patients with Alzheimer's disease. J Neural Transm. 1999;106:499–511.
    1. Parnetti L, Reboldi GP, Gallai V. Cerebrospinal fluid pyruvate levels in Alzheimer's disease and vascular dementia. Neurology. 2000;54:735–737.
    1. Parnetti L, Gaiti A, Brunetti M, Avellini L, Polidori C, Cecchetti R, Palumbo B, Senin U. Increased CSF pyruvate levels as a marker of impaired energy metabolism in Alzheimer's disease. J Am Geriatr Soc. 1995;43:316–318.
    1. Takashima A. GSK-3 is essential in the pathogenesis of Alzheimer's disease. J Alzheimers Dis. 2006;9:309–317.
    1. Baum L, Hansen L, Masliah E, Saitoh T. Glycogen synthase kinase 3 alteration in Alzheimer disease is related to neurofibrillary tangle formation. Mol Chem Neuropathol. 1996;29:253–261.
    1. Peineau S, Taghibiglou C, Bradley C, Wong TP, Liu L, Lu J, Lo E, Wu D, Saule E, Bouschet T, Matthews P, Isaac JT, Bortolotto ZA, Wang YT, Collingridge GL. LTP inhibits LTD in the hippocampus via regulation of GSK3β. Neuron. 2007;53:703–717.
    1. Gomez-Sintes R, Hernandez F, Bortolozzi A, Artigas F, Avila J, Zaratin P, Gotteland JP, Lucas JJ. Neuronal apoptosis and reversible motor deficit in dominant-negative GSK-3 conditional transgenic mice. EMBO J. 2007;26:2743–2754.
    1. Griffin RJ, Moloney A, Kelliher M, Johnston JA, Ravid R, Dockery P, O'Connor R, O'Neill C. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer's disease pathology. J Neurochem. 2005;93:105–117.
    1. Mann DM, Sumpter PQ, Davies CA, Yates PO. Glycogen accumulations in the cerebral cortex in Alzheimer's disease. Acta Neuropathol. 1987;73:181–184.
    1. Liang Z, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Down-regulation of cAMP-dependent protein kinase by over-activated calpain in Alzheimer disease brain. J Neurochem. 2007;103:2462–2470.
    1. Mulder C, Scheltens P, Barkhof F, Gundy C, Verstraeten RA, de Leeuw FE. Low vitamin B6 levels are associated with white matter lesions in Alzheimer's disease. J Am Geriatr Soc. 2005;53:1073–1074.
    1. Mulder C, van der Flier WM, Veerhuis R, Bouwman F, Jakobs C, Verhoeven NM, Barkhof F, Scheltens P, Blankenstein MA. Association between vitamin B6 and white matter hyperintensities in patients with Alzheimer's disease not mediated by homocysteine metabolism. J Am Geriatr Soc. 2007;55:956–958.
    1. Tagliabracci VS, Turnbull J, Wang W, Girard JM, Zhao X, Skurat AV, Delgado-Escueta AV, Minassian BA, Depaoli-Roach AA, Roach PJ. Laforin is a glycogen phosphatase, deficiency of which leads to elevated phosphorylation of glycogen in vivo. Proc Natl Acad Sci USA. 2007;104:19262–19266.
    1. Huang L, Hollingsworth RI, Castellani R, Zipser B. Accumulation of high-molecular-weight amylose in Alzheimer's disease brains. Glycobiology. 2004;14:409–416.
    1. Cross AJ, Crow TJ, Johnson JA, Dawson JM, Peters TJ. Loss of endoplasmic reticulum-associated enzymes in affected brain regions in Huntington's disease and Alzheimer-type dementia. J Neurol Sci. 1985;71:137–143.
    1. Heneka MT, Galea E, Gavriluyk V, Dumitrescu-Ozimek L, Daeschner J, O'Banion MK, Weinberg G, Klockgether T, Feinstein DL. Noradrenergic depletion potentiates β-amyloid-induced cortical inflammation: implications for Alzheimer's disease. J Neurosci. 2002;22:2434–2442.
    1. Nazarali AJ, Reynolds GP. Monoamine neurotransmitters and their metabolites in brain regions in Alzheimer's disease: a postmortem study. Cell Mol Neurobiol. 1992;12:581–587.
    1. Nakamura S, Kawamata T, Akiguchi I, Kameyama M, Kimura H. Reduced nicotinamide adenine dinucleotide phosphate diaphorase histochemistry in neocortex and hippocampus in patients with Alzheimer type dementia and aged controls. Rinsho Shinkeigaku. 1987;27:1059–1063.
    1. Zubenko GS, Moossy J, Claassen D, Martinez AJ, Rao GR. Brain regional analysis of NADH-cytochrome C reductase activity in Alzheimer's disease. J Neuropathol Exp Neurol. 1990;49:206–214.
    1. Jacobs RW, Farivar N, Butcher LL. Alzheimer dementia and reduced nicotinamide adenine dinucleotide (NADH)-diaphorase activity in senile plaques and the basal forebrain. Neurosci Lett. 1985;53:39–44.
    1. Tsuchiya A, Hardy J, Burbacher TM, Faustman EM, Marien K. Fish intake guidelines: incorporating n-3 fatty acid intake and contaminant exposure in the Korean and Japanese communities. Am J Clin Nutr. 2008;87:1867–1875.
    1. Levine KE, Levine MA, Weber FX, Hu Y, Perlmutter J, Grohse PM. Determination of mercury in an assortment of dietary supplements using an inexpensive combustion atomic absorption spectrometry technique. J Automat Meth Manag Chem. 2005;2005:211–216.
    1. Maclean C, Issa A, Newberry S, Mojica W, Morton S, Garland R, Hilton L, Traina S, Shekelle P (2005) Effects of omega-3 fatty acids on cognitive function with aging, dementia, and neurological diseases. Evid Rep Technol Assess (Summ) (114):1–88
    1. Schaefer EJ, Bongard V, Beiser AS, Lamon-Fava S, Robins SJ, Au R, Tucker KL, Kyle DJ, Wilson PW, Wolf PA. Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham Heart Study. Arch Neurol. 2006;63:1545–1550.
    1. Calon F, Lim GP, Yang F, Morihara T, Teter B, Ubeda O, Rostaing P, Triller A, Salem N, Jr, Ashe KH, Frautschy SA, Cole GM. Docosahexaenoic acid protects from dendritic pathology in an Alzheimer's disease mouse model. Neuron. 2004;43:633–645.
    1. Lim GP, Calon F, Morihara T, Yang F, Teter B, Ubeda O, Salem N, Jr, Frautschy SA, Cole GM. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J Neurosci. 2005;25:3032–3040.
    1. Freund-Levi Y, Eriksdotter-Jonhagen M, Cederholm T, Basun H, Faxen-Irving G, Garlind A, Vedin I, Vessby B, Wahlund LO, Palmblad J. Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch Neurol. 2006;63:1402–1408.
    1. Chiu CC, Su KP, Cheng TC, Liu HC, Chang CJ, Dewey ME, Stewart R, Huang SY. The effects of omega-3 fatty acids monotherapy in Alzheimer's disease and mild cognitive impairment: a preliminary randomized double-blind placebo-controlled study. Prog Neuropsychopharmacol Biol Psychiatry. 2008;32:1538–1544.
    1. Scheltens P, Kamphuis PJ, Verhey FR, Olde Rikkert MG, Wurtman RJ, Wilkinson D, Twisk JW, Kurz A. Efficacy of a medical food in mild Alzheimer's disease: a randomized, controlled trial. Alzheimers Dement. 2010;6(1–10):e11.
    1. Cole GM, Frautschy SA. DHA may prevent age-related dementia. J Nutr. 2010;140:869–874.
    1. Cole GM, Ma QL, Frautschy SA. Omega-3 fatty acids and dementia. Prostaglandins Leukot Essent Fatty Acids. 2009;81:213–221.
    1. Heller AR, Rossler S, Litz RJ, Stehr SN, Heller SC, Koch R, Koch T. Omega-3 fatty acids improve the diagnosis-related clinical outcome. Crit Care Med. 2006;34:972–979.
    1. Calon F, Lim GP, Morihara T, Yang F, Ubeda O, Salem NJ, Frautschy SA, G.M. C. Dietary n-3 polyunsaturated fatty acid depletion activates caspases and decreases NMDA receptors in the brain of a transgenic mouse model of Alzheimer's disease. Eur J NeuroSci. 2005;22:617–626.
    1. Green KN, Martinez-Coria H, Khashwji H, Hall EB, Yurko-Mauro KA, Ellis L, LaFerla FM. Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-β and tau pathology via a mechanism involving presenilin 1 levels. J Neurosci. 2007;27:4385–4395.
    1. Oksman M, Iivonen H, Hogyes E, Amtul Z, Penke B, Leenders I, Broersen L, Lutjohann D, Hartmann T, Tanila H. Impact of different saturated fatty acid, polyunsaturated fatty acid and cholesterol containing diets on β-amyloid accumulation in APP/PS1 transgenic mice. Neurobiol Dis. 2006;23:563–572.
    1. Cleland LG, Caughey GE, James MJ, Proudman SM. Reduction of cardiovascular risk factors with longterm fish oil treatment in early rheumatoid arthritis. J Rheumatol. 2006;33:1973–1979.
    1. Little SJ, Lynch MA, Manku M, Nicolaou A. Docosahexaenoic acid-induced changes in phospholipids in cortex of young and aged rats: a lipidomic analysis. Prostaglandins Leukot Essent Fatty Acids. 2007;77:155–162.
    1. Galarraga B, Ho M, Youssef HM, Hill A, McMahon H, Hall C, Ogston S, Nuki G, Belch JJ. Cod liver oil (n-3 fatty acids) as an non-steroidal anti-inflammatory drug sparing agent in rheumatoid arthritis. Rheumatology (Oxford) 2008;47:665–669.
    1. Akbar M, Calderon F, Wen Z, Kim HY. Docosahexaenoic acid: a positive modulator of Akt signaling in neuronal survival. Proc Natl Acad Sci USA. 2005;102:10858–10863.
    1. Rao JS, Ertley RN, Lee HJ, DeMar JC, Jr, Arnold JT, Rapoport SI, Bazinet RP. n-3 polyunsaturated fatty acid deprivation in rats decreases frontal cortex BDNF via a p38 MAPK-dependent mechanism. Mol Psychiatry. 2007;12:36–46.
    1. Wu A, Ying Z, Gomez-Pinilla F. Docosahexaenoic acid dietary supplementation enhances the effects of exercise on synaptic plasticity and cognition. Neuroscience. 2008;155:751–759.
    1. Yavin E, Brand A, Green P. Docosahexaenoic acid abundance in the brain: a biodevice to combat oxidative stress. Nutr Neurosci. 2002;5:149–157.
    1. Hossain MS, Hashimoto M, Gamoh S, Masumura S. Antioxidative effects of docosahexaenoic acid in the cerebrum versus cerebellum and brainstem of aged hypercholesterolemic rats. J Neurochem. 1999;72:1133–1138.
    1. Lukiw WJ, Cui JG, Marcheselli VL, Bodker M, Botkjaer A, Gotlinger K, Serhan CN, Bazan NG. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J Clin Invest. 2005;115:2774–2783.
    1. Innis SM. Dietary (n-3) fatty acids and brain development. J Nutr. 2007;137:855–859.
    1. He C, Qu X, Cui L, Wang J, Kang JX. Improved spatial learning performance of fat-1 mice is associated with enhanced neurogenesis and neuritogenesis by docosahexaenoic acid. Proc Natl Acad Sci USA. 2009;106:11370–11375.
    1. Katakura M, Hashimoto M, Shahdat HM, Gamoh S, Okui T, Matsuzaki K, Shido O. Docosahexaenoic acid promotes neuronal differentiation by regulating basic helix-loop-helix transcription factors and cell cycle in neural stem cells. Neuroscience. 2009;160:651–660.
    1. Pifferi F, Jouin M, Alessandri JM, Haedke U, Roux F, Perriere N, Denis I, Lavialle M, Guesnet P. n-3 Fatty acids modulate brain glucose transport in endothelial cells of the blood-brain barrier. Prostaglandins Leukot Essent Fatty Acids. 2007;77:279–286.
    1. Tsukada H, Kakiuchi T, Fukumoto D, Nishiyama S, Koga K. Docosahexaenoic acid (DHA) improves the age-related impairment of the coupling mechanism between neuronal activation and functional cerebral blood flow response: a PET study in conscious monkeys. Brain Res. 2000;862:180–186.
    1. Hashimoto M, Hossain S, Shimada T, Shido O. Docosahexaenoic acid-induced protective effect against impaired learning in amyloid β-infused rats is associated with increased synaptosomal membrane fluidity. Clin Exp Pharmacol Physiol. 2006;33:934–939.
    1. Hirashima F, Parow AM, Stoll AL, Demopulos CM, Damico KE, Rohan ML, Eskesen JG, Zuo CS, Cohen BM, Renshaw PF. Omega-3 fatty acid treatment and T(2) whole brain relaxation times in bipolar disorder. Am J Psychiatry. 2004;161:1922–1924.
    1. Wassall SR, Stillwell W. Docosahexaenoic acid domains: the ultimate non-raft membrane domain. Chem Phys Lipids. 2008;153:57–63.
    1. Litman BJ, Niu SL, Polozova A, Mitchell DC. The role of docosahexaenoic acid containing phospholipids in modulating G protein-coupled signaling pathways: visual transduction. J Mol Neurosci. 2001;16:237–242.
    1. Salem N, Jr, Litman B, Kim HY, Gawrisch K. Mechanisms of action of docosahexaenoic acid in the nervous system. Lipids. 2001;36:945–959.
    1. de Urquiza AM, Liu S, Sjoberg M, Zetterstrom RH, Griffiths W, Sjovall J, Perlmann T. Docosahexaenoic acid, a ligand for the retinoid X receptor in mouse brain. Science. 2000;290:2140–2144.
    1. Gani OA, Sylte I. Molecular recognition of docosahexaenoic acid by peroxisome proliferator-activated receptors and retinoid-X receptor alpha. J Mol Graph Model. 2008;27:217–224.
    1. Calderon F, Kim HY. Role of RXR in neurite outgrowth induced by docosahexaenoic acid. Prostaglandins Leukot Essent Fatty Acids. 2007;77:227–232.
    1. Ma Q-L, Yang F, Rosario E, Ubeda O, Beech W, Gant D, Chen P, Hudspeth B, Chen C, Zhao Y, Vinters H, Frautschy S, Cole G. Aβ oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via JNK signaling: suppression by omega-3 fatty acids and curcumin. J Neurosci. 2009;29:9078–9089.
    1. Hooijmans CR, Rutters F, Dederen PJ, Gambarota G, Veltien A, van Groen T, Broersen LM, Lutjohann D, Heerschap A, Tanila H, Kiliaan AJ. Changes in cerebral blood volume and amyloid pathology in aged Alzheimer APP/PS1 mice on a docosahexaenoic acid (DHA) diet or cholesterol enriched Typical Western Diet (TWD) Neurobiol Dis. 2007;28:16–29.
    1. Hooijmans CR, Van der Zee CE, Dederen PJ, Brouwer KM, Reijmer YD, van Groen T, Broersen LM, Lutjohann D, Heerschap A, Kiliaan AJ. DHA and cholesterol containing diets influence Alzheimer-like pathology, cognition and cerebral vasculature in APPswe/PS1dE9 mice. Neurobiol Dis. 2009;33:482–498.
    1. Ma QL, Teter B, Ubeda OJ, Morihara T, Dhoot D, Nyby MD, Tuck ML, Frautschy SA, Cole GM. Omega-3 fatty acid docosahexaenoic acid increases SorLA/LR11, a sorting protein with reduced expression in sporadic Alzheimer's disease (AD): relevance to AD prevention. J Neurosci. 2007;27:14299–14307.
    1. Puskas LG, Kitajka K, Nyakas C, Barcelo-Coblijn G, Farkas T. Short-term administration of omega 3 fatty acids from fish oil results in increased transthyretin transcription in old rat hippocampus. Proc Natl Acad Sci USA. 2003;100:1580–1585.
    1. Zhao L, Teter B, Morihara T, Lim GP, Ambegaokar SS, Ubeda OJ, Frautschy SA, Cole GM. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer's disease intervention. J Neurosci. 2004;24:11120–11126.
    1. Nourooz-Zadeh J, Liu EHC, Yhlen B, Anggard EE, Halliwell B. F4-Isoprostanes as specific marker of docosahexaenoic acid peroxidation in Alzheimer's disease. J Neurochem. 1999;72:734–740.
    1. Montine KS, Quinn JF, Zhang J, Fessel JP, Roberts LJ, 2nd, Morrow JD, Montine TJ. Isoprostanes and related products of lipid peroxidation in neurodegenerative diseases. Chem Phys Lipids. 2004;128:117–124.
    1. Cole GM, Frautschy SA. Docosahexaenoic acid protects from amyloid and dendritic pathology in an Alzheimer's disease mouse model. Nutr Health. 2006;18:249–259.
    1. Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold. Adv Exp Med Biol. 2007;595:1–75.
    1. Cole GM, Teter B, Frautschy SA. Neuroprotective effects of curcumin. Adv Exp Med Biol. 2007;595:197–212.
    1. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci. 2001;21:8370–8377.
    1. Cole GM, Yang F, Lim GP, Cummings JL, Masterman DL, Frautschy SA. A rationale for curcuminoids for the prevention or treatment of Alzheimer's disease. Curr Med Chem Immunol Endocr Metab Agents. 2003;3:15–25.
    1. Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar SS, Chen PP, Kayed R, Glabe CG, Frautschy SA, Cole GM. Curcumin inhibits formation of amyloid β oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J Biol Chem. 2005;280:5892–5901.
    1. Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ. Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. J Neurochem. 2007;102:1095–1104.
    1. Ryu EK, Choe YS, Lee KH, Choi Y, Kim BT. Curcumin and dehydrozingerone derivatives: synthesis, radiolabeling, and evaluation for β-amyloid plaque imaging. J Med Chem. 2006;49:6111–6119.
    1. Caughey B, Raymond LD, Raymond GJ, Maxson L, Silveira J, Baron GS. Inhibition of protease-resistant prion protein accumulation in vitro by curcumin. J Virol. 2003;77:5499–5502.
    1. Brunden KR, Ballatore C, Crowe A, Smith AB 3rd, Lee VM, Trojanowski JQ (2009) Tau-directed drug discovery for Alzheimer's disease and related tauopathies: a focus on tau assembly inhibitors. Exp Neurol (in press)
    1. Narlawar R, Pickhardt M, Leuchtenberger S, Baumann K, Krause S, Dyrks T, Weggen S, Mandelkow E, Schmidt B. Curcumin-derived pyrazoles and isoxazoles: Swiss army knives or blunt tools for Alzheimer's disease? ChemMedChem. 2008;3:165–172.
    1. Frautschy SA, Hu W, Kim P, Miller SA, Chu T, Harris-White ME, Cole GM. Phenolic anti-inflammatory antioxidant reversal of Aβ-induced cognitive deficits and neuropathology. Neurobiol Aging. 2001;22:993–1005.
    1. Huang MT, Lysz T, Ferraro T, Abidi TF, Laskin JD, Conney AH. Inhibitory effects of curcumin on in vitro lipoxygenase and cyclooxygenase activities in mouse epidermis. Cancer Res. 1991;51:813–819.
    1. Hong J, Bose M, Ju J, Ryu JH, Chen X, Sang S, Lee MJ, Yang CS. Modulation of arachidonic acid metabolism by curcumin and related β-diketone derivatives: effects on cytosolic phospholipase A(2), cyclooxygenases and 5-lipoxygenase. Carcinogenesis. 2004;25:1671–1679.
    1. Kelloff GJ, Crowell JA, Hawk ET, Steele VE, Lubet RA, Boone CW, Covey JM, Doody LA, Omenn GS, Greenwald P, Hong WK, Parkinson DR, Bagheri D, Baxter GT, Blunden M, Doeltz MK, Eisenhauer KM, Johnson K, Knapp GG, Longfellow DG, Malone WF, Nayfield SG, Seifried HE, Swall LM, Sigman CC. Strategy and planning for chemopreventive drug development: clinical development plan: curcumin. J Cell Biochem Suppl. 1996;26:72–85.
    1. Rao CV, Rivenson A, Simi B, Reddy BS. Chemoprevention of colon carcinogenesis by dietary curcumin, a naturally occurring plant phenolic compound. Cancer Res. 1995;55:259–266.
    1. Ikonomovic MD, Abrahamson EE, Uz T, Manev H, Dekosky ST. Increased 5-lipoxygenase immunoreactivity in the hippocampus of patients with Alzheimer's disease. J Histochem Cytochem. 2008;56:1065–1073.
    1. Firuzi O, Zhuo J, Chinnici CM, Wisniewski T, Pratico D. 5-Lipoxygenase gene disruption reduces amyloid-β pathology in a mouse model of Alzheimer's disease. FASEB J. 2008;22:1169–1178.
    1. Chen YR, Tan TH. Inhibition of the c-Jun N-terminal kinase (JNK) signaling pathway by curcumin. Oncogene. 1998;17:173–178.
    1. Gao M, Zhou H, Li X. Curcumin protects PC12 cells from corticosterone-induced cytotoxicity: possible involvement of the ERK1/2 pathway. Basic Clin Pharmacol Toxicol. 2009;104:236–240.
    1. Yao Y, Chinnici C, Tang H, Trojanowski JQ, Lee VM, Pratico D. Brain inflammation and oxidative stress in a transgenic mouse model of Alzheimer-like brain amyloidosis. J Neuroinflammation. 2004;1:21.
    1. Andorfer C, Acker CM, Kress Y, Hof PR, Duff K, Davies P. Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms. J Neurosci. 2005;25:5446–5454.
    1. Polydoro M, Acker CM, Duff K, Castillo PE, Davies P. Age-dependent impairment of cognitive and synaptic function in the htau mouse model of tau pathology. J Neurosci. 2009;29:10741–10749.
    1. Tabaton M. Oxidative stress and β-APP proteolytic processing. Neurobiol Aging. 2004;25(S2):S69 (S64-02-03).
    1. Baum L, Ng A. Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models. J Alzheimers Dis. 2004;6:367–377.
    1. Begum AN, Jones MR, Lim GP, Morihara T, Kim P, Heath DD, Rock CL, Pruitt MA, Yang F, Hudspeth B, Hu S, Faull KF, Teter B, Cole GM, Frautschy SA. Curcumin structure-function, bioavailability, and efficacy in models of neuroinflammation and Alzheimer's disease. J Pharmacol Exp Ther. 2008;326:196–208.
    1. Kang SK, Cha SH, Jeon HG. Curcumin-induced histone hypoacetylation enhances caspase-3-dependent glioma cell death and neurogenesis of neural progenitor cells. Stem Cells Dev. 2006;15:165–174.
    1. Kim SJ, Son TG, Park HR, Park M, Kim MS, Kim HS, Chung HY, Mattson MP, Lee J. Curcumin stimulates proliferation of embryonic neural progenitor cells and neurogenesis in the adult hippocampus. J Biol Chem. 2008;283:14497–14505.
    1. Rao P, Knaus EE. Evolution of nonsteroidal anti-inflammatory drugs (NSAIDs): cyclooxygenase (COX) inhibition and beyond. J Pharm Pharm Sci. 2008;11:81s–110s.
    1. Baum L, Lam CW, Cheung SK, Kwok T, Lui V, Tsoh J, Lam L, Leung V, Hui E, Ng C, Woo J, Chiu HF, Goggins WB, Zee BC, Cheng KF, Fong CY, Wong A, Mok H, Chow MS, Ho PC, Ip SP, Ho CS, Yu XW, Lai CY, Chan MH, Szeto S, Chan IH, Mok V. Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol. 2008;28:110–113.
    1. Garcea G, Jones DJ, Singh R, Dennison AR, Farmer PB, Sharma RA, Steward WP, Gescher AJ, Berry DP. Detection of curcumin and its metabolites in hepatic tissue and portal blood of patients following oral administration. Br J Cancer. 2004;90:1011–1015.
    1. Gota VS, Maru GB, Soni TG, Gandhi TR, Kochar N, Agarwal MG. Safety and pharmacokinetics of a solid lipid curcumin particle formulation in osteosarcoma patients and healthy volunteers. J Agric Food Chem. 2010;58:2095–2099.

Source: PubMed

3
订阅