mitochondrial abnormalities: a primary basis for oxidative damage in alzheimer's disease

8
© 1999 Wiley-Liss, Inc. DRUG DEVELOPMENT RESEARCH 46:26–33 (1999) Research Overview Mitochondrial Abnormalities: A Primary Basis for Oxidative Damage in Alzheimer’s Disease Mark A. Smith, 1 * Keisuke Hirai, 1 Akihiko Nunomura, 1,2 and George Perry 1 1 Institute of Pathology, Case Western Reserve University, Cleveland, Ohio 2 Department of Psychiatry and Neurology, Asahikawa Medical College, Asahikawa, Japan ABSTRACT The most striking feature of Alzheimer’s disease (AD) is the number of abnormalities affecting essentially every aspect of brain homeostasis. Recent work suggests that increased oxidative stress that damages lipids, proteins, and nucleic acids and results in the accumulation of redox-active metals may be responsible for the diversity of systems involved. Interestingly, all of the genetic factors, β- protein precursor, presenilins, and apolipoprotein E, have been linked to reactive oxygen species produc- tion or apoptosis, a process intimately associated with oxidative stress. This leaves open the question of why oxidative damage is increased in AD. In studies of mitochondria, we demonstrated increased mito- chondrial DNA specifically in vulnerable neurons in cases of AD, suggesting that AD is marked by a fundamental abnormality in neuronal metabolism. Oxidative stress, therefore, seems to be the element linking the multitude of changes in Alzheimer’s disease to a fundamental metabolic deficiency. Drug Dev. Res. 46:26–33, 1999. © 1999 Wiley-Liss, Inc. Key words: Alzheimer’s disease; oxidative damage; mitochondrial abnormalities Strategy, Management and Health Policy Venture Capital Enabling Technology Preclinical Research Preclinical Development Toxicology, Formulation Drug Delivery, Pharmacokinetics Clinical Development Phases I-III Regulatory, Quality, Manufacturing Postmarketing Phase IV INTRODUCTION Damage by reactive oxygen is not only found in the proteins of neurofibrillary tangles (NFTs) and se- nile plaques of Alzheimer’s disease (AD) but also in the cytoplasm of neuronal populations vulnerable to death during the course of AD [Smith et al., 1994a,b, 1995a–c, 1996a,b, 1997a,b; Sayre et al., 1997a]. To understand the significance of reactive oxygen dam- age, investigators have been studying two crucial as- pects: (1) the relationship between oxidative stress and other factors, established by genetic or epidemiologic studies to play an important role in AD; and (2) the metabolic basis for increased reactive oxygen damage. Here, we present evidence that oxidative damage is a central process in neurodegeneration in AD that links together the diverse factors important for disease ini- tiation and progression. Indeed, not only does oxida- tive damage occur before formation of the pathologic lesions, thereby making it one of the earliest cyto- pathologic markers of neuronal dysfunction in AD, but it further serves as a marker to uncover the underlying origin of AD, which may be a metabolic disorder. AD ALTERS ESSENTIALLY EVERY FACET OF BRAIN HOMEOSTASIS: AGING AND OXIDATIVE STRESS The pathologic presentation of AD, the leading cause of senile dementia, involves regionalized neuronal death and an accumulation of intraneuronal and extracellular lesions termed NFTs and senile plaques, respectively [re- viewed in Smith, 1998]. Several independent hypotheses have been proposed to link the pathologic lesions and neu- ronal cytopathology with, among others, apolipoprotein E genotype [Corder et al., 1993; Roses, 1995], hyperpho- sphorylation of cytoskeletal proteins [Trojanowski et al., Grant sponsor: National Institutes of Health; Grant number: AG09287; Grant sponsor: American Health Assistance Foundation (AHAF); Grant sponsor: Alzheimer’s Association. *Correspondence to: Mark A. Smith, Institute of Pathology, Case Western Reserve University, 2085 Adelbert Road, Cleveland, OH. E-mail: [email protected]

Upload: mark-a-smith

Post on 06-Jun-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

26 SMITH ET AL.

© 1999 Wiley-Liss, Inc.

DRUG DEVELOPMENT RESEARCH 46:26–33 (1999)

Research Overview

Mitochondrial Abnormalities: A Primary Basis forOxidative Damage in Alzheimer’s Disease

Mark A. Smith,1* Keisuke Hirai,1 Akihiko Nunomura,1,2 and George Perry1

1Institute of Pathology, Case Western Reserve University, Cleveland, Ohio2Department of Psychiatry and Neurology, Asahikawa Medical College, Asahikawa, Japan

ABSTRACT The most striking feature of Alzheimer’s disease (AD) is the number of abnormalitiesaffecting essentially every aspect of brain homeostasis. Recent work suggests that increased oxidativestress that damages lipids, proteins, and nucleic acids and results in the accumulation of redox-activemetals may be responsible for the diversity of systems involved. Interestingly, all of the genetic factors, β-protein precursor, presenilins, and apolipoprotein E, have been linked to reactive oxygen species produc-tion or apoptosis, a process intimately associated with oxidative stress. This leaves open the question ofwhy oxidative damage is increased in AD. In studies of mitochondria, we demonstrated increased mito-chondrial DNA specifically in vulnerable neurons in cases of AD, suggesting that AD is marked by afundamental abnormality in neuronal metabolism. Oxidative stress, therefore, seems to be the elementlinking the multitude of changes in Alzheimer’s disease to a fundamental metabolic deficiency. Drug Dev.Res. 46:26–33, 1999. © 1999 Wiley-Liss, Inc.

Key words: Alzheimer’s disease; oxidative damage; mitochondrial abnormalities

Strategy, Management and Health Policy

Venture CapitalEnablingTechnology

PreclinicalResearch

Preclinical DevelopmentToxicology, FormulationDrug Delivery,Pharmacokinetics

Clinical DevelopmentPhases I-IIIRegulatory, Quality,Manufacturing

PostmarketingPhase IV

INTRODUCTION

Damage by reactive oxygen is not only found inthe proteins of neurofibrillary tangles (NFTs) and se-nile plaques of Alzheimer’s disease (AD) but also inthe cytoplasm of neuronal populations vulnerable todeath during the course of AD [Smith et al., 1994a,b,1995a–c, 1996a,b, 1997a,b; Sayre et al., 1997a]. Tounderstand the significance of reactive oxygen dam-age, investigators have been studying two crucial as-pects: (1) the relationship between oxidative stress andother factors, established by genetic or epidemiologicstudies to play an important role in AD; and (2) themetabolic basis for increased reactive oxygen damage.Here, we present evidence that oxidative damage is acentral process in neurodegeneration in AD that linkstogether the diverse factors important for disease ini-tiation and progression. Indeed, not only does oxida-tive damage occur before formation of the pathologiclesions, thereby making it one of the earliest cyto-pathologic markers of neuronal dysfunction in AD, but

it further serves as a marker to uncover the underlyingorigin of AD, which may be a metabolic disorder.

AD ALTERS ESSENTIALLY EVERY FACET OF BRAINHOMEOSTASIS: AGING AND OXIDATIVE STRESS

The pathologic presentation of AD, the leading causeof senile dementia, involves regionalized neuronal deathand an accumulation of intraneuronal and extracellularlesions termed NFTs and senile plaques, respectively [re-viewed in Smith, 1998]. Several independent hypotheseshave been proposed to link the pathologic lesions and neu-ronal cytopathology with, among others, apolipoprotein Egenotype [Corder et al., 1993; Roses, 1995], hyperpho-sphorylation of cytoskeletal proteins [Trojanowski et al.,

Grant sponsor: National Institutes of Health; Grant number:AG09287; Grant sponsor: American Health Assistance Foundation(AHAF); Grant sponsor: Alzheimer’s Association.

*Correspondence to: Mark A. Smith, Institute of Pathology,Case Western Reserve University, 2085 Adelbert Road, Cleveland,OH. E-mail: [email protected]

Page 2: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

MITOCHONDRIAL ABNORMALITIES IN ALZHEIMER’S DISEASE 27

1993a], and amyloid-β metabolism [Selkoe, 1997]. How-ever, not one of these theories alone is sufficient to explainthe diversity of abnormalities found in AD that involves amultitude of cellular and biochemical changes. Further-more, attempts to mimic AD by a perturbation of one ofthese elements using cell or animal models, includingtransgenic animals, do not result in the same spectrum ofpathologic alterations. Perhaps the most striking exampleof this is that while amyloid-β plaques are deposited insome transgenic rodent models overexpressing β-proteinprecursor, there is no neuronal loss [Irizarry et al.,1997a,b]—a seminal feature of AD. This diversity ofchanges suggests that AD stems from a shift in neuronalmetabolism, paralleling the key feature of AD, aging.

AD is a disease with a prevalence that increasesexponentially with aging [Katzman, 1986] (Fig. 1). Im-portantly, this holds true even in individuals with a ge-netic predisposition, i.e., those individuals with anautosomal dominant inheritance of AD or in individualswith Down’s syndrome who develop the pathology of ADin middle age. The free radical theory of aging [Harman,1956] hypothesizes that the aging process is associatedwith multisystem failure due to oxidative damage causedby an imbalance between reactive oxygen production andantioxidant defenses. A corollary of this theory is thataging results in compromised ability to deal with patho-logic sources of reactive oxygen. Furthermore, the abil-ity of reactive oxygen to modify lipid proteins and nucleicacids leaves cells open to multiple system failure or, morecorrectly for those surviving cells, multiple compensa-tions to maintain homeostatic balance.

OXIDATIVE DAMAGE AND RESPONSE

An exact determination of the contribution of eachsource of reactive oxygen is complicated, if for no otherreason than that most sources have positive feedback.Nonetheless, the overall result of unchecked oxygen radi-cals is damage. Such damage found in AD includes ad-vanced glycation end-products [Smith et al., 1994a;Ledesma et al., 1994; Vitek et al., 1994; Yan et al., 1994],

nitration [Good et al., 1996; Smith et al., 1997a], lipidperoxidation adduction products [Montine et al., 1996a;Sayre et al., 1997a], as well as carbonyl-modifiedneurofilament protein and free carbonyls [Smith et al.,1991, 1995b, 1996a]. Importantly, this damage involvesall neurons in populations vulnerable to death in AD, notjust those containing NFTs. What is particularly strikingis that the neurons displaying oxidative damage show noovert signs of degeneration.

The cytopathologic significance of oxidative dam-age is seen by the up-regulation of antioxidant enzymessuch as heme oxygenase-1 [Smith et al., 1994b; Schipperet al., 1995; Premkumar et al., 1995] and superoxidedismutase-1 [Pappolla et al., 1992] in neurons with NFTs.Furthermore, neurons with NFTs accumulate dimethyl-argininase [Smith et al., 1998a], a regulator whose enzy-matic activity removes methylarginine-related inhibitionof nitric oxide synthetase, thus increasing nitric oxide pro-duction. Because nitric oxide reacts with superoxide toform peroxynitrite, increased nitric oxide can be seen asa possible response to increased reactive oxygen. Quan-titative immunocytochemical studies of cases of AD showthat there is a complete overlap between neurons up-regulating heme oxygenase-1 and Alz50, an early markerof τ abnormalities, indicating that cytoskeletal abnormali-ties are associated with increased oxidative stress or viceversa [Smith and Perry, unpublished observation].

PHOSPHORYLATION AS A RESPONSE TOOXIDATIVE STRESS

The mechanisms by which normal soluble cyto-skeletal elements such as τ and neurofilaments are trans-formed into insoluble paired helical filaments is animportant issue [Selkoe et al., 1982; Smith et al., 1996b].Insolubility has been linked to abnormal phosphoryla-tion, the most well known posttranslational change of τ[Goedert et al., 1991; Greenberg et al., 1992], and a num-ber of specific kinases and phosphates have been impli-cated [reviewed in Trojanowski et al., 1993a]. However,while increased phosphorylation decreases microtubulestability, a salient feature of the pathology of AD [Perryet al., 1991; Alonso et al., 1994, 1996; Iqbal et al., 1994;Praprotnik et al., 1996a,b], NFT insolubility is not medi-ated by phosphorylation [Smith et al., 1996b]. Indeed, invitro phosphorylation of normal τ or complete dephos-phorylation of NFT has no effect on their solubility[Goedert et al., 1991; Greenberg et al., 1992; Gustke etal., 1992; Smith et al., 1996b]. Moreover, recent studiessuggest the τ phosphorylation found in AD may be partof a novel process similar to that seen during mitosis [Popeet al., 1994; Preuss et al., 1995], suggesting that neuronsaffected in the disease might be abortively entering thecell cycle [Vincent et al., 1996; McShea et al., 1997, 1999].

Phosphorylation is intimately tied to oxidative stressFig. 1. The prevalence of Alzheimer’s disease is strictly age dependent.Based on the 1998 United States Government Accounting Office report.

Page 3: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

28 SMITH ET AL.

by the extracellular receptor kinase (ERK) pathway[Guyton et al., 1996] and by activation of the transcrip-tion factor NFκB [Schreck et al., 1991]. Although thereis controversy concerning the kinases involved in thephosphorylation of τ in AD, the ERK pathway is impli-cated [Ledesma et al., 1992; Trojanowski et al., 1993b;Hyman et al., 1994]. In studies with antibodies specificto activated ERK, we found activity in all pyramidal hip-pocampal neurons in cases of AD but none in controls(Fig. 2). Therefore, abnormal phosphorylation of proteinsin AD may not only be a consequence of oxidative stressbut also a protective response that neurons use to evadethe onset of apoptotic death brought on by oxidativestress. The increased oxidative damage in vulnerableneurons in AD brings neurons to a point of decidingwhether to accept oxidative death through apoptosis,marked by the JNK pathway [Gardner and Johnson,1996], or instead, enter the ERK pathway.

REACTIVE OXYGEN LINKS GENETICS TO ADPATHOGENESIS

A number of mechanisms have been suggested forthe neurotoxicity of amyloid-β [Yankner et al., 1990; re-viewed in Iversen et al., 1995; Sayre et al., 1997b], in-cluding membrane depolarization [Carette et al., 1993],increased sensitivity to excitotoxins [Koh et al., 1990], andalterations in calcium homeostasis [Mattson et al., 1992];

however, the influences of amyloid-β and other geneticfactors on AD may be through their effect on oxidativestress (Fig. 3). Indeed, the leading hypothesis is that neu-ronal damage by amyloid-β is mediated by free radicalsand, as such, can be attenuated using antioxidants suchas vitamin E [Behl et al., 1992, 1994] or catalase [Lockhartet al., 1994; Zhang et al., 1996]. Furthermore, amyloid-βis reported to spontaneously generate peptidyl radicals[Butterfield et al., 1994; Goodman et al., 1994; Harris etal., 1995; Prehn et al., 1996], and mutations in β-precur-sor protein are associated with increased DNA fragmen-tation, possibly involving oxidative mechanisms (seebelow). Critically addressing whether amyloid-β initiatesoxidative damage in AD requires careful study of the re-lationship of amyloid-β deposition and increased oxida-tive damage. In this regard, it is extremely interestingthat the recently reported transgenic rodent models ofamyloid-β deposition [Games et al., 1995; Hsiao et al.,1996] show the full spectrum of oxidative changes of AD[Smith et al., 1998b; Pappolla et al., 1998].

Presenilins 1 and 2 [Sherrington et al., 1995; Selkoe,1997] are genetic factors in which the biological mecha-nism, although not established, may also involve oxida-tive damage. Increased presenilin 2 expression increasesDNA fragmentation and apoptotic changes [Wolozin etal., 1996], both important consequences of oxidative dam-age. Apolipoprotein E, in brains and cerebrospinal fluid,is found adducted with the highly reactive lipidperoxidation product, hydroxynonenal [Montine et al.,1996b]. Furthermore, apolipoprotein E is a strong chela-tor of copper and iron, important redox-active transitionmetals [Miyata and Smith, 1996]. Finally, interaction ofapolipoprotein E with amyloid-β only occurs in the pres-ence of oxygen [Strittmatter et al., 1993].

APOPTOSIS

In programmed cell death, i.e., apoptosis, cells aredigested within their own membrane by proteases andnucleases as well as by increased reactive oxygen. How-

Fig. 2. Hypothetical time line of the alterations in neuronal structure inAlzheimer’s disease (AD). With the onset of AD, normal neurons developnumerous forms of oxidative damage, including nitration (nitrotyrosine),protein oxidation (free carbonyls), and lipid peroxide adducts (lipidperoxidation) prior to the formation of neurofibrillary tangles (pre-NFTs)that evolved into intracellular (I-NFT) and, consequenctly, extracellular(E-NFT) NFTs. Coincident with oxidative damage, extracellular receptorkinase (ERK) is activated. These findings suggest that phosphorylation ab-normalities may be a result of oxidative damage.

Fig. 3. The primary epidemiologic and genetic factors implicated inAlzheimer’s disease are directly linked to oxidative stress.

Page 4: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

MITOCHONDRIAL ABNORMALITIES IN ALZHEIMER’S DISEASE 29

ever, without the full range of morphologic changes, it isunclear whether DNA fragmentation is apoptotic or is,instead, mediated solely by oxidative damage to DNAand resulting repair [Berlin and Haseltine, 1981]. Therelative contribution of oxidative stress-related versusapoptosis-related DNA fragmentation in AD is unre-solved. Yet, bearing on this issue, the relative infrequencyof apoptosis defined by morphology [Su et al., 1994;Cotman and Su, 1996] and the broad findings of frag-mentation in all cells in cases of AD argues for wide-spread oxidative DNA damage rather than widespreadapoptosis. Certainly, this interpretation is consistent withthe oxidative nuclear damage in all cells of the brain inareas affected in AD [Smith et al., 1996a, 1997b].

WHAT IS THE INITIAL SOURCE OF INCREASEDREACTIVE OXYGEN PRODUCTION IN AD?

Reactive oxygen is a ubiquitous byproduct of bothoxidative phosphorylation and the myriad of oxidases nec-essary to support aerobic metabolism. In AD, in additionto this background level of reactive oxygen, there are anumber of additional contributory sources that are thoughtto play an important role in the disease process: (1) Iron,in a redox-active state, is increased in NFTs as well as inamyloid-β deposits [Good et al., 1992; Smith et al., 1997b].Iron catalyzes the formation of ·OH from H2O2 as well asthe formation of advanced glycation end-products. Fur-thermore, aluminum, which also accumulates in neu-rofibrillary tangle-containing neurons [Good et al., 1992],stimulates iron-induced lipid peroxidation [Oteiza, 1994].(2) Activated microglia, such as those that surround mostsenile plaques [Cras et al., 1990], are a source of NO andO2

–· [Colton and Gilbert, 1987], which can react to formperoxynitrite, leaving nitrotyrosine as an identifiablemarker [Good et al., 1996; Smith et al., 1997a]. (3) Amy-loid-β itself has been directly implicated in reactive oxy-gen formation through peptidyl radicals [Butterfield et al.,1994; Hensley et al., 1994; Sayre et al., 1997b]. (4) Ad-vanced glycation end-products in the presence of transi-tion metals (see above) can undergo redox cycling withconsequent reactive oxygen species production [Baynes,1991; Yan et al., 1994, 1995]. Additionally, advancedglycation end-products, as well as amyloid-β, activate spe-cific receptors such as the receptor for advanced glycationend-products (RAGE) and the class A scavenger-receptorto increase reactive oxygen production [Yan et al., 1996;El Khoury et al., 1996]. (5) Abnormalities in the mitochon-drial genome [Corral-Debrinski et al., 1994; Davis et al.,1997] or deficiencies in key metabolic enzymes [Sorbi etal., 1983; Sheu et al., 1985; Sims et al., 1987; Blass et al.,1990; Parker et al., 1990] suggest that metabolic abnor-malities affecting mitochondria may be the major and pos-sible initiating source of reactive oxygen in AD.

Although there is support for all listed sources of re-

active oxygen, with the exception of the last, mitochondrialabnormalities, they should only be operative after forma-tion of senile plaques and NFTs. Yet as stated earlier, neu-ronal oxidative damage occurs in the absence of NFTs oradjacent senile plaques. In consideration of the low diffu-sion of reactive oxygen through tissue, these observationspoint to a source within neuronal soma, again supportingmitochondria. Although while there has been considerablecontroversy regarding the role of mitochondrial inheritancein AD [Davis et al., 1997; Hirano et al., 1997; Hutchin et al.,1997; Wallace et al., 1997], we took the view that if mito-chondria are important in increased reactive oxygen gen-eration, mitochondria should show abnormalities in AD. Toassess this issue, we performed in situ hybridization witholigonucleotide probes recognizing wild-type as well asdamaged mitochondrial DNA (mtDNA), i.e., with the 4977-kb common deletion. We found that vulnerable neurons,but no other neurons or glia, showed striking increases inmtDNA [Hirai et al., 1998a,b]. These findings support reac-tive oxygen from mitochondria as the initial source of oxida-tive damage in AD and the earliest biochemical abnormality.Whether the biochemical abnormality is reflected cytologi-cally as increased mitochondria requires further ultrastruc-tural study of neurons in AD cases compared withage-matched controls. Unfortunately, all historical studiesof AD compared neurons with NFTs with those lacking themand presumed neurons lacking NFTs were unaffected bythe disease. Our results invalidate that assumption and showthat all vulnerable neurons are affected.

Although we do not know the reason for mtDNAincrease in AD, its restriction to large neurons suggeststhe increase might be linked to the energy demands ofneurons. That the increased mtDNA is only found in casesof AD, where amyloid-β is also increased, and that amy-loid-β affects a decrease in mitochondrial potential [Prehnet al., 1996] leaves open the possibility that amyloid-βbased mitochondrial damage may be at the heart of thematter. Further studies explicitly addressing the relation-ships between increased mtDNA and amyloid-β depos-its in normal aging and AD are clearly required.

THERAPEUTICS

Oxidative damage resulting, totally or in part, frommitochondrial abnormalities is the earliest cytopathologicand biochemical change of AD. Therefore, it is not sur-prising that agents that inhibit free radical formation[Schinetti et al., 1987; Hamburger and McCay, 1990;Jones et al., 1995; Rong et al., 1996; Wu et al., 1996; Behlet al., 1997; Igawa et al., 1997; Neuzil et al., 1997; Rippleet al., 1997; Shoda et al., 1997; Vane and Botting, 1997]reduce both the incidence and the progression of AD(Fig. 4) [McGeer and Rogers, 1992; Rogers et al., 1993;Breitner et al., 1994; Münch et al., 1994, 1997; Rich etal., 1995; Colaco et al., 1996; Kanowski et al., 1996;

Page 5: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

30 SMITH ET AL.

Smalheiser and Swanson, 1996; Stoll et al., 1996; Thal etal., 1996; Henderson, 1997; Kawas et al., 1997;Papasozomenos, 1997; Sano et al., 1997; Shoda et al.,1997; Skolnick, 1997; Stewart et al., 1997]. This relation-ship, together with the efficacy of metal chelation treat-ment [McLachlan et al., 1991], strongly suggest thatoxidative stress is a central process in AD and that agentsthat prevent oxidative damage will be particularly effica-cious in the treatment for the disease.

REFERENCESAlonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. 1994. Role of abnor-

mally phosphorylated tau in the breakdown of microtubules inAlzheimer’s disease. Proc Natl Acad Sci USA 91:5562–5566.

Alonso AC, Grundke-Iqbal I, Iqbal K. 1996. Alzheimer’s diseasehyperphosphorylated tau sequesters normal tau into tangles of fila-ments and disassembles microtubules. Nat Med 2:783–787.

Baynes JW. 1991. Role of oxidative stress in development of complica-tions in diabetes. Diabetes 40:405–412.

Behl C, Davis J, Cole GM, Schubert D. 1992. Vitamin E protects nervecells from amyloid-β protein toxicity. Biochem Biophys Res Commun186:944–950.

Behl C, Davis JB, Lesley R, Schubert D. 1994. Hydrogen peroxidemediates amyloid-β protein toxicity. Cell 77:817–827.

Behl C, Skutella T, Lezoualc’h F, Post A, Widmann M, Newton CJ,Holsboer F. 1997. Neuroprotection against oxidative stress by es-trogens: structure-activity relationship. Mol Pharmacol 51:535–541.

Berlin V, Haseltine WA. 1981. Reduction of adriamycin to a semi-quinone-free radical by NADPH cytochrome P-450 reductase pro-duces DNA cleavage in a reaction mediated by molecular oxygen. JBiol Chem 256:4747–4756.

Blass JP, Baker AC, Ko L, Black RS. 1990. Induction of Alzheimerantigens by an uncoupler of oxidative phosphorylation. Arch Neurol47:864–869.

Breitner JCS, Gau BA, Welsh KA, Plassman BL, McDonald WM,Helms MJ, Anthony J. 1994. Inverse association of anti-inflamma-tory treatments and Alzheimer’s disease: initial results of a co-twincontrol study. Neurology 44:227–232.

Butterfield DA, Hensley K, Harris M, Mattson M, Carney J. 1994. β-amyloid peptide free radical fragments initiate synaptosomallipoperoxidation in a sequence-specific fashion: implications toAlzheimer’s disease. Biochem Biophys Res Commun 200:710–715.

Carette B, Poulain P, Delacourte A. 1993. Electrophysiological effects

of 25-35 amyloid-β-protein on guinea-pig lateral septal neurons.Neurosci Lett 151:111–114.

Colaco CA, Ledesma MD, Harrington CR, Avila J. 1996. The role ofthe Maillard reaction in other pathologies: Alzheimer’s disease.Nephrol Dial Transplant 11(Suppl 5):7–12.

Colton CA, Gilbert DL. 1987. Production of superoxide anions by aCNS macrophage, the microglia. FEBS Lett 223:284–288.

Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, GaskellPC, Small GW, Roses AD, Haines JL, Pericak-Vance MA. 1993. Genedose of apolipoprotein E type 4 allele and the risk of Alzheimer’sdisease in late onset families. Science 261:921–923.

Corral-Debrinski M, Horton T, Lott MT, Shoffner JM, McKee AC,Beal MF, Graham BH, Wallace DC. 1994. Marked changes in mito-chondrial DNA deletion levels in Alzheimer brains. Genomics23:471–476.

Cotman CW, Su JH. 1996. Mechanisms of neuronal death inAlzheimer’s disease. Brain Pathol 6:493–506.

Cras P, Kawai M, Siedlak S, Mulvihill P, Gambetti P, Lowery D,Gonzalez-DeWhitt P, Greenberg B, Perry G. 1990. Neuronal andmicroglial involvement in β-amyloid protein deposition inAlzheimer’s disease. Am J Pathol 137:241–246.

Davis RE, Miller S, Herrnstadt C, Ghosh SS, Fahy E, Shinobu LA,Galasko D, Thal LJ, Beal MF, Howell N, Parker WD Jr. 1997. Muta-tions in mitochondrial cytochrome c oxidase genes segregate withlate-onset Alzheimer disease. Proc Natl Acad Sci USA94:4526–4531.

El Khoury J, Hickman SE, Thomas CA, Cao L, Silverstein SC, LoikeJD. 1996. Scavenger receptor-mediated adhesion of microglia to β-amyloid fibrils. Nature 382:716–719.

Games D, Adams D, Alessandrini R, Barbour R, Berthelette P,Blackwell C, Carr T, Clemens J, Donaldson T, Gillespie F, GuidoT, Hagopian S, Johnson-Wood K, Khan K, Lee M, Leibowitz P,Lieberburg I, Little S, Masliah E, McConlogue L, Montoya-Zavala M, Mucke L, Paganini L, Penniman E, Power M, SchenkD, Seubert P, Snyer B, Soriano F, Tan H, Vitale J, Wadsworth S,Wolozin B, Zhao J. 1995. Alzheimer-type neuropathology intransgenic mice overexpressing V717F β-amyloid precursorprotein. Nature 373:523–527.

Gardner AM, Johnson GL. 1996. Fibroblast growth factor-2 suppres-sion of tumor necrosis factor alpha-mediated apoptosis requires Rasand the activation of mitogen-activated protein kinase. J Biol Chem271:14560–14566.

Goedert M, Sisodia SS, Price DL. 1991. Neurofibrillary tangles andbeta-amyloid deposits in Alzheimer’s disease. Curr Opin Neurobiol1:441–447.

Good PF, Perl DP, Bierer LM, Schmeidler J. 1992. Selective accumu-lation of aluminum and iron in the neurofibrillary tangles ofAlzheimer’s disease: a laser microprobe (LAMMA) study. AnnNeurol 31:286–292.

Good PF, Werner P, Hsu A, Olanow CW, Perl DP. 1996. Evidence ofneuronal oxidative damage in Alzheimer’s disease. Am J Pathol149:21–28.

Goodman Y, Steiner MR, Steiner SM, Mattson MP. 1994. Nordi-hydroguaiaretic acid protects hippocampal neurons against amy-loid beta-peptide toxicity, and attenuates free radical and calciumaccumulation. Brain Res 654:171–176.

Greenberg SG, Davies P, Schein JD, Binder LI. 1992. Hydrofluoricacid-treated tau PHF proteins display the same biochemical prop-erties as normal tau. J Biol Chem 267:564–569.

Gustke N, Steiner B, Mandelkow EM, Biernat J, Meyer HE, GoedertM, Mandelkow E. 1992. The Alzheimer-like phosphorylation of tau

Fig. 4. All known efficacious agents either affecting incidence or pro-gression of Alzheimer’s disease have antioxidant activity.

Page 6: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

MITOCHONDRIAL ABNORMALITIES IN ALZHEIMER’S DISEASE 31

protein reduces microtubule binding and involves Ser-Pro and Thr-Pro motifs. FEBS Lett 307:199–205.

Guyton KZ, Liu Y, Gorospe M, Xu Q, Holbrook NJ. 1996. Activation ofmitogen-activated protein kinase by H2O2: role in cell survival fol-lowing oxidant injury. J Biol Chem 271:4138–4142.

Hamburger SA, McCay PB. 1990. Spin trapping of ibuprofen radicals:evidence that ibuprofen is a hydroxyl radical scavenger. Free RadicRes Commun 9:337–342.

Harman D. 1956. Ageing: a theory based on free radical and radiationchemistry. J Gerontol 11:298–300.

Harris ME, Carney JM, Cole PS, Hensley K, Howard BJ, Martin L,Bummer P, Wang Y, Pedigo NWJ, Butterfield DA. 1995. β-amyloidpeptide-derived, oxygen-dependent free radicals inhibit glutamateuptake in cultured astrocytes: implications for Alzheimer’s disease.Neuroreport 6:1875–1879.

Henderson VW. 1997. The epidemiology of estrogen replacementtherapy and Alzheimer’s disease. Neurology 48(Suppl 7):S27–S35.

Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF,Floyd RA, Butterfield DA. 1994. A model for β-amyloid aggrega-tion and neurotoxicity based on free radical generation by the pep-tide: relevance to Alzheimer disease. Proc Natl Acad Sci USA91:3270–3274.

Hirai K, Smith MA, Wade R, Perry G. 1998a. Vulnerable neurons inAlzheimer disease accumulate mitochondrial DNA with the com-mon 5kb deletion. J Neuropathol Exp Neurol 57:511.

Hirai K, Smith MA, Wade R, Perry G. 1998b. Neuronal RNA oxida-tion and mitochondrial proliferation denote profound metabolicabnormalities in Alzheimer disease. Neurobiol Aging 19 (Suppl45):54–55.

Hirano M, Shtilbans A, Mayeux R, Davidson MM, DiMauro S, KnowlesJA, Schon EA. 1997. Apparent mtDNA heteroplasmy in Alzheimer’sdisease patients and in normals due to PCR amplification of nucleus-embedded mtDNA pseudogenes. Proc Natl Acad Sci USA 94:14894–14899.

Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S,Yang F, Cole G. 1996. Correlative memory deficits, Aβ elevation,and amyloid plaques in transgenic mice. Science 274:99–102.

Hutchin TP, Heath PR, Pearson RC, Sinclair AJ. 1997. MitochondrialDNA mutations in Alzheimer’s disease. Biochem Biophys ResCommun 241:221–225.

Hyman BT, Elvhage TE, Reiter J. 1994. Extracellular signal regulatedkinases: localization of protein and mRNA in the human hippocam-pal formation in Alzheimer’s disease. Am J Pathol 144:565–572.

Igawa K, Maruyama R, Katayama I, Nishioka K. 1997. Anti-oxidativetherapy with oral dapsone improved HCV antibody positive annu-lar elastolytic giant cell granuloma. J Dermatol 24:328–331.

Iqbal K, Zaidi T, Bancher C, Grundke-Iqbal I. 1994. Alzheimer pairedhelical filaments: restoration of the biological activity by dephos-phorylation. FEBS Lett 349:104–108.

Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT. 1997a.APPSw transgenic mice develop age-related A beta deposits andneuropil abnormalities, but no neuronal loss in CA1. J NeuropatholExp Neurol 56:965–973.

Irizarry MC, Soriano F, McNamara M, Page KJ, Schenk D, Games D,Hyman BT. 1997b. Aβ deposition is associated with neuropil changes,but not with overt neuronal loss in the human amyloid precursorprotein V717F (PDAPP) transgenic mouse. J Neurosci 17:7053–7059.

Iversen LL, Mortishire-Smith RJ, Pollack SJ, Shearman MS. 1995. Thetoxicity in vitro of β-amyloid protein. Biochem J 331:1–16.

Jones LH, Abdalla DS, Freitas JC. 1995. Effects of indole-3-acetic acidon croton oil- and arachidonic acid-induced mouse ear edema.Inflamm Res 44:372–375.

Kanowski S, Herrmann WM, Stephan K, Wierich W, Horr R. 1996.Proof of efficacy of the Ginkgo biloba special extract EGb 761 inoutpatients suffering from mild to moderate primary degenerativedementia of the Alzheimer type or multi-infarct dementia.Pharmacopsychiatry 29:47–56.

Katzman R. 1986. Alzheimer’s disease. N Engl J Med 314:964–973.

Kawas C, Resnick S, Morrison A, Brookmeyer R, Corrada M,Zonderman A, Bacal C, Lingle DD, Metter E. 1997. A prospectivestudy of estrogen replacement therapy and the risk of developingAlzheimer’s disease: the Baltimore Longitudinal Study of Aging.Neurology 48:1517–1521.

Koh JY, Yang LL, Cotman CW. 1990. β-amyloid protein increases thevulnerability of cultured cortical neurons to excitotoxic damage. BrainRes 533:315–320.

Ledesma MD, Correas I, Avila J, Diaz-Nido J. 1992. Implication ofbrain cdc2 and MAP2 kinases in the phosphorylation of tau proteinin Alzheimer’s disease. FEBS Lett 308:218–224.

Ledesma MD, Bonay P, Colaco C, Avila J. 1994. Analysis of microtu-bule-associated protein tau glycation in paired helical filaments. JBiol Chem 269:21614–21619.

Lockhart BP, Benicourt C, Junien JL, Privat A. 1994. Inhibitors of freeradical formation fail to attenuate direct β-amyloid25-35 peptide-mediated neurotoxicity in rat hippocampal cultures. J Neurosci Res39:494–505.

Mattson MP, Cheng B, Davis D, Bryant K, Lieberburg I, Rydel RE.1992. β-amyloid peptides destabilize calcium homeostasis and ren-der human cortical neurons vulnerable to excitotoxicity. J Neurosci12:376–389.

McGeer PL, Rogers J. 1992. Anti-inflammatory agents as a therapeu-tic approach to Alzheimer’s disease. Neurology 42:447–449.

McLachlan DR, Kruck TP, Lukiw WJ, Krishnan SS. 1991. Would de-creased aluminum ingestion reduce the incidence of Alzheimer’sdisease? Can Med Assoc 145:793–804.

McShea A, Harris PLR, Webster KR, Wahl A, Smith MA. 1997. Ab-normal expression of the cell cycle regulators P16 and CDK4 inAlzheimer’s disease. Am J Pathol 150:1933–1939.

McShea A, Wahl AF, Smith MA. 1999. Re-entry into the cell cycle: amechanism for neurodegeneration in Alzheimer disease. Med Hy-potheses, in press.

Miyata M, Smith JD. 1996. Apolipoprotein E allele-specific antioxi-dant activity and effects on cytotoxicity by oxidative insults and β-amyloid peptides. Nat Genet 14:55–61.

Montine TJ, Amarnath V, Martin ME, Strittmatter WJ, Graham DG.1996a. E-4-hydroxy-2-nonenal is cytotoxic and cross-links cytoskeletalproteins in P19 neuroglial cultures. Am J Pathol 148:89–93.

Montine TJ, Huang DY, Valentine WM, Amarnath V, Saunders A,Weisgraber KH, Graham DG, Strittmatter WJ. 1996b. Crosslinkingof apolipoprotein E by products of lipid peroxidation. J NeuropatholExp Neurol 55:202–210.

Münch G, Taneli Y, Schraven E, Schindler U, Schinzel R, Palm D,Riederer P. 1994. The cognition-enhancing drug tenilsetam is aninhibitor of protein crosslinking by advanced glycosylation. J Neu-ral Trans-Parkinsons Dis Dem Sect 8:193–208.

Münch G, Mayer S, Michaelis J, Hipkiss AR, Riederer P, Muller R,Neumann A, Schinzel R, Cunningham AM. 1997. Influence of ad-vanced glycation end-products and AGE-inhibitors on nucleation-

Page 7: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

32 SMITH ET AL.

dependent polymerization of beta-amyloid peptide. BiochimBiophys Acta 1360:17–29.

Neuzil J, Witting PK, Stocker R. 1997. Alpha-tocopheryl hydro-quinone is an efficient multifunctional inhibitor of radical-initi-ated oxidation of low density lipoprotein lipids. Proc Natl AcadSci USA 94:7885–7890.

Oteiza PI. 1994. A mechanism for the stimulatory effect of aluminum oniron-induced lipid peroxidation. Arch Biochem Biophys 308:374–379.

Papasozomenos SC. 1997. The heat shock-induced hyperphos-phorylation of tau is estrogen-independent and prevented by an-drogens: implications for Alzheimer disease. Proc Natl Acad Sci USA94:6612–6617.

Pappolla MA, Omar RA, Kim KS, Robakis NK. 1992. Immunohis-tochemical evidence of oxidative stress in Alzheimer’s disease. AmJ Pathol 140:621–628.

Pappolla M, Chyan Y-J, Omar RA, Hsiao K, Perry G, Smith MA, BoznerP. 1998. Evidence of oxidative stress and in vivo neurotoxicity of β-amyloid in a transgenic mouse model of Alzheimer’s disease: achronic oxidative paradigm for testing antioxidant therapies in vivo.Am J Pathol 152:871–877.

Parker WD Jr, Filley CM, Parks JK. 1990. Cytochrome oxidative defi-ciency in Alzheimer’s disease. Neurology 40:1302–1303.

Perry G, Kawai M. Tabaton M, Onorato M, Mulvihill P, Richey P,Morandi A, Connolly JA, Gambetti P. 1991. Neuropil threads ofAlzheimer’s disease show a marked alteration of the normal cyto-skeleton. J Neurosci 11:1748–1755.

Pope WB, Lambert MP, Leypold B, Seupaul R, Sletten L, Krafft G,Klein WL. 1994. Microtubule-associated protein tau is hyper-phosphorylated during mitosis in the human neuroblastoma cell lineSH-SY5Y. Exp Neurol 126:185–194.

Praprotnik D, Smith MA, Richey PL, Vinters HV, Perry G. 1996a.Plasma membrane fragility in dystrophic neurites in senile plaquesof Alzheimer’s disease: an index of oxidative stress. Acta Neuropathol91:1–5.

Praprotnik D, Smith MA, Richey PL, Vinters HV, Perry G. 1996b. Fila-ment heterogeneity within the dystrophic neurites of senile plaquessuggests blockage of fast axonal transport in Alzheimer’s disease.Acta Neuropathol 91:226–235.

Prehn JH, Bindokas VP, Jordan J, Galindo MF, Ghadge GD, RoosRP, Boise LH, Thompson CB, Krajewski S, Reed JC, Miller RJ.1996. Protective effect of transforming growth factor-β1 on β-amyloid neurotoxicity in rat hippocampal neurons. Mol Pharm-acol 49:319–328.

Premkumar DRD, Smith MA, Richey PL, Petersen RB, Castellani R,Kutty RK, Wiggert B, Perry G, Kalaria RN. 1995. Induction of hemeoxygenase-1 mRNA and protein in neocortex and cerebral vesselsin Alzheimer’s disease. J Neurochem 65:1399–1402.

Preuss U, Doring F, Illenberger S, Mandelkow EM. 1995. Cell cycle-dependent phosphorylation and microtubule binding of tau proteinstably transfected into Chinese hamster ovary cells. Mol Biol Cell6:1397–1410.

Rich JB, Rasmusson DX, Folstein MF, Carson KA, Kawas C, Brandt J.1995. Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease.Neurology 45:51–55.

Ripple MO, Henry WF, Rago RP, Wilding G. 1997. Prooxidant-anti-oxidant shift induced by androgen treatment of human prostate car-cinoma cells. J Natl Cancer Inst 89:40–48.

Rogers J, Kirby LC, Hempelman SR, Berry DL, McGeer PL, KaszniakAW, Zalinski J, Cofield M, Mansukhani L, Willson P, Kogan F. 1993.Clinical trial of indomethacin in Alzheimer’s disease. Neurology43:1609–1611.

Rong Y, Geng Z, Lau BH. 1996. Ginkgo biloba attenuates oxidativestress in macrophages and endothelial cells. Free Radic Biol Med20:121–127.

Roses AD. 1995. On the metabolism of apolipoprotein E and the Alzhei-mer diseases. Exp Neurol 132:149–156.

Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, GrundmanM, Woodbury P, Growdon J, Cotman CW, Pfeiffer E, Schneider LS,Thal LJ. 1997. A controlled trial of selegiline, alpha-tocopherol, orboth as treatment for Alzheimer’s disease. The Alzheimer’s DiseaseCooperative Study. N Engl J Med 336:1216–1222.

Sayre LM, Zelasko DA, Harris PLR, Perry G, Salomon RG, SmithMA. 1997a. 4-Hydroxynonenal-derived advanced lipid peroxidationend products are increased in Alzheimer’s disease. J Neurochem68:2092–2097.

Sayre LM, Zagorski MG, Surewicz WK, Krafft GA, Perry G. 1997b.Mechanisms of neurotoxicity associated with amyloid β depositionand the role of free radicals in the pathogenesis of Alzheimer’s dis-ease: a critical appraisal. Chem Res Toxicol 10:518–526.

Schinetti ML, Rossini D, Greco R, Bertelli A. 1987. Protective actionof acetylcarnitine on NADPH-induced lipid peroxidation of cardiacmicrosomes. Drugs Exp Clin Res 13:509–515.

Schipper HM, Cisse S, Stopa EG. 1995. Expression of heme oxygen-ase-1 in the senescent and Alzhemer-diseased brain. Ann Neurol37:758–768.

Schreck R, Rieber P, Baeuerle PA. 1991. Reactive oxygen intermedi-ates as apparently widely used messengers in the activation of theNF-κB transcription factor and HIV-1. EMBO J 10:2247–2258.

Selkoe DJ. 1997. Alzheimer’s disease: genotypes, phenotypes, andtreatments. Science 275:630–631.

Selkoe DJ, Ihara Y, Salazar FJ. 1982. Alzheimer’s disease: insolubilityof partially purified paired helical filaments in sodium dodecyl sul-fate and urea. Science 215:1243–1245.

Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, IkedaM, Chi H, Lin C, Li G, Holman K, Tsuda T, Mar L, Foncin J-F,Bruni AC, Montesi MP, Sorbi S, Rainero I, Pinessi L, Polinsky RJ,Wasco W, Da Silva HAR, Haines JL, Pericak-Vance MA, Tanzi RE,Roses AD, Fraser PE, Rommens JM, St. George-Hyslop PH. 1995.Cloning of a gene bearing missense mutations in early-onset famil-ial Alzheimer’s disease. Nature 375:754–760.

Sheu KF, Kim YT, Blass JP, Weksler ME. 1985. An immunochemicalstudy of the pyruvate dehydrogenase deficit in Alzheimer’s diseasebrain. Ann Neurol 17:444–449.

Shoda H, Miyata S, Liu BF, Yamada H, Ohara T, Suzuki K, Oimomi M,Kasuga M. 1997. Inhibitory effects of tenilsetam on the Maillardreaction. Endocrinology 138:1886–1892.

Sims NR, Finegan JM, Blass JP, Bowen DM, Neary D. 1987. Mito-chondrial function in brain tissue in primary degenerative demen-tia. Brain Res 436:30–38.

Skolnick AA. 1997. Old Chinese herbal medicine used for fever yieldspossible new Alzheimer disease therapy. J Am Med Assoc 277:776.

Smalheiser NR, Swanson DR. 1996. Indomethacin and Alzheimer’sdisease. Neurology 46:583.

Smith MA. 1998. Alzheimer disease. In: Bradley JY, Harris RA, edi-tors. International Review of Neurobiology. San Diego: AcademicPress. p 1–54.

Smith CD, Carney JM, Starke-Reed PE, Oliver CN, Stadtman ER,Floyd RA, Marksberry WR. 1991. Excess brain protein oxidativeand enzyme dysfunction in normal aging and in Alzheimer disease.Proc Natl Acad Sci USA 88:10540–10543.

Page 8: Mitochondrial abnormalities: A primary basis for oxidative damage in Alzheimer's disease

MITOCHONDRIAL ABNORMALITIES IN ALZHEIMER’S DISEASE 33

Smith MA, Taneda S, Richey PL, Miyata S, Yan S-D, Stern D, SayreLM, Monnier VM, Perry G. 1994a. Advanced Maillard reaction prod-ucts are associated with Alzheimer disease pathology. Proc Natl AcadSci USA 91:5710–5714.

Smith MA, Kutty RK, Richey PL, Yan S-D, Stern D, Chader GJ, WiggertB, Petersen RB, Perry G. 1994b. Heme oxygenase-1 is associatedwith the neurofibrillary pathology of Alzheimer’s disease. Am JPathol 145:42–47.

Smith MA, Sayre LM, Monnier VM, Perry G. 1995a. Radical AGEingin Alzheimer’s disease. Trends Neurosci 18:172–176.

Smith MA, Rudnicka-Nawrot M, Richey PL, Praprotnik D, MulvihillP, Miller CA, Sayre LM, Perry G. 1995b. Carbonyl-related post-translational modification of neurofilament protein in the neurofibril-lary pathology of Alzheimer’s disease. J Neurochem 64:2660–2666.

Smith MA, Sayre LM, Vitek MP, Monnier VM, Perry G. 1995c. EarlyAGEing and Alzheimer’s. Nature 374:316.

Smith MA, Perry G, Richey PL, Sayre LM, Anderson VE, BealMF, Kowall N. 1996a. Oxidative damage in Alzheimer’s. Na-ture 382:120–121.

Smith MA, Siedlak SL, Richey PL, Nagaraj RH, Elhammer A,Perry G. 1996b. Quantitative solubilization and analysis of in-soluble paired helical filaments from Alzheimer disease. BrainRes 717:99–108.

Smith MA, Harris PLR, Sayre LM, Beckman JS, Perry G. 1997a. Wide-spread peroxynitrite-mediated damage in Alzheimer’s disease. JNeurosci 17:2653–2657.

Smith MA, Harris PLR, Sayre LM, Perry G. 1997b. Iron accumulationin Alzheimer disease is a source of redox-generated free radicals.Proc Natl Acad Sci USA 94:9866–9868.

Smith MA, Vašák M, Knipp M, Castellani RJ, Perry G. 1998a.Dimethylargininase, a nitric oxide regulatory protein, in Alzheimerdisease. Free Radic Biol Med 25:898–902.

Smith MA, Hirai K, Hsiao K, Pappolla MA, Harris PLR, Siedlak SL,Tabaton M, Perry G. 1998b. Amyloid-β deposition in Alzheimertransgenic mice initiates oxidative stress. J Neurochem 70:2212–2215.

Sorbi S, Bird ED, Blass JP. 1983. Decreased pyruvate dehydrogenasecomplex activity in Huntington and Alzheimer brain. Ann Neurol13:72–78.

Stewart WF, Kawas C, Corrada M, Metter EJ. 1997. Risk of Alzheimer’sdisease and duration of NSAID use. Neurology 48:626–631.

Stoll S, Scheuer K, Pohl O, Muller WE. 1996. Ginkgo biloba extract(EGb 761) independently improves changes in passive avoidancelearning and brain membrane fluidity in the aging mouse.Pharmacopsychiatry 29:144–149.

Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS,Pericak-Vance M, Schmechel D, Saunders AM, Goldgaber D, RosesAD. 1993. Binding of human apolipoprotein E to synthetic amyloidβ peptide: isoform-specific effects and implications for late-onsetAlzheimer disease. Proc Natl Acad Sci USA 90:8098–8102.

Su JH, Anderson AJ, Cummings BJ, Cotman CW. 1994. Immunohis-tochemical evidence for apoptosis in Alzheimer’s disease. Neuroreport5:2529–2533.

Thal LJ, Carta A, Clarke WR, Ferris SH, Friedland RP, Petersen RC,Pettegrew JW, Pfeiffer E, Raskind MA, Sano M, Tuszynski MH,Woolson RF. 1996. A 1-year multicenter placebo-controlled studyof acetyl-L-carnitine in patients with Alzheimer’s disease. Neurol-ogy 47:705–711.

Trojanowski JQ, Schmidt ML, Shin R-W, Bramblett GT, Goedert M,Lee VM-Y. 1993a. PHF-τ (A68): from pathological marker to poten-tial mediator of neuronal dysfunction and degeneration inAlzheimer’s disease. Clin Neurosci 1:184–191.

Trojanowski JQ, Mawal-Dewan M, Schmidt ML, Martin J, Lee VM.1993b. Localization of the mitogen activated protein kinase ERK2in Alzheimer’s disease neurofibrillary tangles and senile plaqueneurites. Brain Res 618:333–337.

Vane JR, Botting RM. 1997. Mechanism of action of aspirin-like drugs.Semin Arthritis Rheum 26(6 Suppl 1):2–10.

Vincent I, Rosado M, Davies P. 1996. Mitotic mechanisms inAlzheimer’s disease? J Cell Biol 132:413–425.

Vitek MP, Bhattacharya K, Glendening JM, Stopa E, Vlassara H, BucalaR, Manogue K, Cerami A. 1994. Advanced glycation end productscontribute to amyloidosis in Alzheimer disease. Proc Natl Acad SciUSA 91:4766–4770.

Wallace DC, Stugard C, Murdock D, Schurr T, Brown MD. 1997. An-cient mtDNA sequences in the human nuclear genome: a potentialsource of errors in identifying pathogenic mutations. Proc Natl AcadSci USA 94:14900–14905.

Wolozin B, Iwasaki K, Vito P, Ganjei JK, Lacana E, Sunderland T, ZhaoB, Kusiak JW, Wasco W, D’Adamio L. 1996. Participation of presenilin2 in apoptosis: enhanced basal activity conferred by an Alzheimermutation. Science 274:1710–1713.

Wu RM, Murphy DL, Chiueh CC. 1996. Suppression of hydroxyl radi-cal formation and protection of nigral neurons by 1-deprenyl(selegiline). Ann NY Acad Sci 786:379–390.

Yan S-D, Chen X, Schmidt A-M, Brett J, Godman G, Zou Y-S, ScottCW, Caputo C, Frappier T, Smith MA, Perry G, Yen S-H, Stern D.1994. Glycated tau protein in Alzheimer disease: a mechanism forinduction of oxidant stress. Proc Natl Acad Sci USA 91:7787–7791.

Yan SD, Yan SF, Chen X, Fu J, Chen M, Kuppusamy P, Smith MA,Perry G, Godman GC, Nawroth P, Zweier JL, Stern D. 1995. Non-enzymatically glycated tau in Alzheimer’s disease induces neuronaloxidant stress resulting in cytokine gene expression and release ofamyloid β-peptide. Nat Med 1:693–699.

Yan SD, Chen X, Fu J, Chen M, Zhu H, Roher A, Slattery T, Zhao L,Nagashima M, Morser J, Migheli A, Nawroth P, Stern D, SchmidtAM. 1996. RAGE and amyloid-β peptide neurotoxicity inAlzheimer’s disease. Nature 382:685–691.

Yankner BA, Duffy LK, Kirschner DA. 1990. Neurotrophic and neu-rotoxic effects of amyloid beta protein: reversal by tachykinin neu-ropeptides. Science 250:279–282.

Zhang Z, Rydel RE, Drzewiecki GJ, Fuson K, Wright S, WogulisM, Audia JE, May PC, Hyslop PA. 1996. Amyloid beta-mediatedoxidative and metabolic stress in rat cortical neurons: no directevidence for a role for H2O2 generation. J Neurochem 67:1595–1606.