Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

17
doi:10.1152/physiol.00024.2010 26:54-69, 2011. Physiology Paul H. Axelsen, Hiroaki Komatsu and Ian V. J. Murray Pathogenesis of Alzheimer's Disease Oxidative Stress and Cell Membranes in the You might find this additional info useful... 363 articles, 102 of which can be accessed free at: This article cites http://physiologyonline.physiology.org/content/26/1/54.full.html#ref-list-1 1 other HighWire hosted articles This article has been cited by [PDF] [Full Text] [Abstract] , January 2, 2012; 287 (1): 748-756. J. Biol. Chem. Yi-Jiong Zhang, Jing-Ming Shi, Cai-Juan Bai, Han Wang, Hai-Yun Li, Yi Wu and Shang-Rong Ji Peptide Are Competing Processes as a Result of Distinct Patterns of Motif Interplay β Intra-membrane Oligomerization and Extra-membrane Oligomerization of Amyloid- including high resolution figures, can be found at: Updated information and services http://physiologyonline.physiology.org/content/26/1/54.full.html can be found at: Physiology about Additional material and information http://www.the-aps.org/publications/physiol This infomation is current as of January 24, 2012. ESSN: 1548-9221. Visit our website at http://www.the-aps.org/. Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2011 by the American Physiological Society. ISSN: 1548-9213, developments. It is published bimonthly in February, April, June, August, October, and December by the American Physiological (formerly published as News in Physiological Science) publishes brief review articles on major physiological Physiology on January 24, 2012 physiologyonline.physiology.org Downloaded from

Transcript of Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

Page 1: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

doi:10.1152/physiol.00024.2010 26:54-69, 2011.PhysiologyPaul H. Axelsen, Hiroaki Komatsu and Ian V. J. MurrayPathogenesis of Alzheimer's DiseaseOxidative Stress and Cell Membranes in the

You might find this additional info useful...

363 articles, 102 of which can be accessed free at:This article cites http://physiologyonline.physiology.org/content/26/1/54.full.html#ref-list-1

1 other HighWire hosted articlesThis article has been cited by

  [PDF] [Full Text] [Abstract]

, January 2, 2012; 287 (1): 748-756.J. Biol. Chem.Yi-Jiong Zhang, Jing-Ming Shi, Cai-Juan Bai, Han Wang, Hai-Yun Li, Yi Wu and Shang-Rong JiPeptide Are Competing Processes as a Result of Distinct Patterns of Motif Interplay

βIntra-membrane Oligomerization and Extra-membrane Oligomerization of Amyloid-

including high resolution figures, can be found at:Updated information and services http://physiologyonline.physiology.org/content/26/1/54.full.html

can be found at:Physiologyabout Additional material and information http://www.the-aps.org/publications/physiol

This infomation is current as of January 24, 2012. 

ESSN: 1548-9221. Visit our website at http://www.the-aps.org/.Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2011 by the American Physiological Society. ISSN: 1548-9213,developments. It is published bimonthly in February, April, June, August, October, and December by the American Physiological

(formerly published as News in Physiological Science) publishes brief review articles on major physiologicalPhysiology

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 2: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

Oxidative Stress and Cell Membranes inthe Pathogenesis of Alzheimer’s Disease

Amyloid � proteins and oxidative stress are believed to have central roles in

the development of Alzheimer’s disease. Lipid membranes are among the

most vulnerable cellular components to oxidative stress, and membranes in

susceptible regions of the brain are compositionally distinct from those in

other tissues. This review considers the evidence that membranes are either a

source of neurotoxic lipid oxidation products or the target of pathogenic

processes involving amyloid � proteins that cause permeability changes or ion

channel formation. Progress toward a comprehensive theory of Alzheimer’s

disease pathogenesis is discussed in which lipid membranes assume both

roles and promote the conversion of monomeric amyloid � proteins into

fibrils, the pathognomonic histopathological lesion of the disease.

Paul H. Axelsen,1,2 Hiroaki Komatsu,1and Ian V. J. Murray3

Departments of 1Pharmacology, 2Biochemistry and Biophysics,and Medicine, University of Pennsylvania School of Medicine,Philadelphia, Pennsylvania; and 3Department of Neuroscience

and Experimental Therapeutics, Texas A&M Health ScienceCenter, College Station, Texas

The literature of Alzheimer’s disease (AD) researchis vast, complex, and often contradictory. Never-theless, a broad perspective on this literature, andon what typically does and does not happen inbiological systems, suggests either that lipid mem-brane damage is directly involved in the pathogen-esis of AD or that it is an important consequence ofAD. Therefore, investigations into the relationshipbetween membrane damage and amyloidogenesismay yield important insights into AD pathogenesis.

The “amyloid hypothesis” has driven much ofthe research on AD pathogenesis since it was pro-posed in 1991 (123, 124). In its simplest form, thishypothesis suggests that the accumulation of am-yloid beta (A�) proteins in brain tissue drives thepathogenesis of AD. A� proteins originate from alarge transmembrane protein of unknown functionknown as amyloid precursor protein (APP) by theaction of �-secretase and �-secretase activity(FIGURE 1). The specific cleavage site of �-secre-tase is somewhat variable, yielding proteins rang-ing from 39 to 43 residues in length, with 40 and 42residue forms predominating (A�40 and A�42).The non-amyloidogenic pathway for APP process-ing involves �-secretases, a family of enzymes thatcleave APP near the middle of the A� proteinsegment.

The amyloid hypothesis was initially based ongenetic studies of familial AD and the occurrenceof AD-like pathology in Down Syndrome. Amongthe most cited experimental studies in support ofthis hypothesis are those in which A� proteinsimpair physiological and cognitive function wheninjected into rodent brains (72, 136, 184, 282, 320,336). Nevertheless, the amyloid hypothesis hasbeen criticized for being incomplete and vague.

For example, it is unable to explain nonfamilial orsporadic cases of AD (the most prevalent form),how fibril formation leads to neuronal death, thepoor clinicopathological correlation between am-yloid plaque burden and cognitive impairment(279), the hyperphosphorylation and aggregationof tau proteins as neurofibrillary tangles, and ax-onopathies that precede amyloid deposition (302).These criticisms notwithstanding, there are no al-ternative hypotheses with nearly as much support,and the amyloid hypothesis is readily adapted toanswer some of these criticisms. For example, thepathologies that precede amyloid deposition maybe due to prefibrillar intermediate forms of A�

protein, and plaques may represent an inert formof the protein (62, 125, 337). Supporting this idea isthe observation that a variant form of A� that oli-gomerizes but does not fibrillize can nonethelesscause several pathological manifestations of ADother than fibril formation (318).

It has been suggested that there is a tenuousbalance among the rates of A� protein production,aggregation, and elimination in brain tissue, suchthat a small disturbance applied over sufficienttime causes the pathological accumulation of A�

protein as amyloid fibrils in AD. However, the con-centrations of A� proteins in the brain appear to befar lower than their aqueous in vitro solubility (143,238, 281). Therefore, A� proteins should not formamyloid fibrils in the brain even if concentrationsare modestly elevated. Yet, fibrils do form withadvancing age in virtually everyone, indicating thatfactors other than A� protein concentration musthave a role in causing them to form. If smallchanges in protein concentration cannot accountfor fibril formation, then we must consider factors

REVIEWSPHYSIOLOGY 26: 54–69, 2011; doi:10.1152/physiol.00024.2010

1548-9213/11 ©2011 Int. Union Physiol. Sci./Am. Physiol. Soc.54

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 3: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

such as macromolecular crowding (216, 228),locally high concentrations of A� proteins at syn-apses (71), chemical modification (30, 161, 231,287, 360), cross-linking (17, 22, 290, 291), or metalcomplex formation (6, 31, 92, 105, 153, 272, 284,306). Understanding the chemical mechanismsthat reduce the solubility of A� proteins in braintissue is key to understanding why AD pathologydevelops in some individuals and not others.

Oxidative Stress

Oxidative stress is caused by a dense, complex andheterogeneous network of oxidizing reactions runningcounter to the reducing conditions that otherwise pre-vail in cells and tissues. It has been suggested that theaccumulation of A� proteins in the brain may be aprotective response to oxidative stress (16, 28, 162, 164,273, 292). The density of plaques containing A� proteincorrelates inversely with markers of oxidative damage(75), and the cortical deposition of A� proteinscorrelates with reduced oxidative damage inDown syndrome (236). Moreover, A� proteins pre-vent lipoprotein oxidation (163) and metal-inducedneuronal death in culture (363). However, it is inher-ently difficult to quantify oxidative stress, and attemptsto do this are subject to many methodological pitfalls.For example, a study concluding that the primarymechanism of A� toxicity does not involve oxidativepathways measured thiobarbituric acid reactive sub-stances (TBARS) to assess lipid peroxidation (248).However, assays for TBARS measure only a small sub-set of the diverse oxidation products generated, as wellas substances not derived from lipids. Other studiessuggesting that A� has antioxidant activity were notcontrolled for the inherent redox activity of peptidegroups or conducted in the presence of biologicallyrelevant electron donors and acceptors to drive thereactions (23, 24).

Overall, it seems more likely that A� proteinspromote oxidative stress (42, 50, 91, 201, 202, 254).The brain in AD appears to sustain more oxidativedamage than normal brain (200, 211, 331), exhibitsan increased susceptibility to oxidative stress (190,220, 277), and has relatively low levels of naturallyoccurring antioxidants such as �-tocopherol (144,358). Regions of the brain rich in A� proteins alsohave increased levels of protein oxidation (129).The overexpression of A� proteins in transgenicmice, in C. elegans, and in cell culture, increasesbiomarkers of oxidative stress (256, 345). A� pro-teins cause H2O2 and lipid peroxides to accumu-late in cells (27). Catalase protects cells from A�

toxicity, and cell lines selected for resistance to A�

toxicity also become resistant to the cytotoxic ac-tion of H2O2. A� proteins promote the oxidation ofcompounds such as dopamine, phospholipids, andcholesterol (40, 134, 135, 235, 241, 257, 356) as long

as an intact methionine side chain is present (5, 21,45, 47, 49, 232).

A� proteins and amyloid plaques bind redox-active transition metals that are likely to be theactual site of redox activity (76, 273). Copper levelsare significantly increased in amyloid plaques (183,191, 304), although comparatively subtle increasesin copper transport across cell membranes maycause significant changes in A� protein turnover(321). Excess dietary copper increases AD-like pa-thology in a mouse model (160). Cu2� and Zn2�

chelators appear to inhibit A� deposition in thebrains of mouse models (64), and metal depletionpromotes the disaggregation of A� fibrils (65).Cu2� potentiates the neurotoxicity of A�42 � A�40in embryonic rodent neurons, and its effect is me-diated by H2O2 (135, 355). Studies of copper in thepathogenesis of AD have been extensively reviewed(39, 90, 100, 305). In contrast, Zn2� ions are redox-inert and able to protect/rescue human cells intissue culture from A� and Cu2� toxicity (75). Arecent study, however, observed that the zinc re-leased at synapses with neurotransmitters causedthe accumulation of toxic A� oligomers to thesemembranes (86).

It has been suggested that A� proteins split intofragments that are both neurotoxic and able togenerate additional oxygen radicals (48, 128), al-though these findings have been strongly refuted(89). Nevertheless, electron paramagnetic reso-nance spectroscopy has shown that there is astrong correlation between the intensity of radicalgeneration by A� and neurotoxicity (219). In thesestudies, preincubation of A� to form fibrils in-creased its toxicity. In contrast, replacing the re-dox-active sulfur atom in residue Met35 withmethylene (CH2) resulted in a peptide that formedfibrillar structures but had no demonstrable toxic-ity toward cultured hippocampal neurons. In thesame experimental system, vitamin E (presumablyacting as an antioxidant) neutralized the neurotox-icity of A� but had no effect on its ability to form

FIGURE 1. Processing of the amyloid precursor proteinA� proteins are 39- to 43-residue segments within the 770-residue amyloid precursorprotein (APP), beginning at residue 672. The non-amyloidogenic processing pathway iscatalyzed by �-secretase, which cleaves in the midst of the A� protein segment. Theamyloidogenic pathway is catalyzed by �-secretase, which cleaves an extracellular site ofAPP, and �-secretase, which cleaves at points within the transmembrane segment.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 55

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 4: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

fibrils (332). Another study concluding that freeradicals do not mediate A�-induced neurotoxicityused ginkgolides (a purported anti-oxidant compo-nent of Ginkgo biloba leaves) and vitamin E toinhibit oxidation (351). Although neither agentprotected cells from apoptosis and death, it is un-likely that they completely halted radical-mediatedoxidative damage.

Proteomics studies of oxidative stress have fo-cused on proteins damaged by oxidation, nitration,and other reactive substances (41, 44, 50, 51, 57, 58,60, 80, 107, 243, 285, 293, 317). In most cases,reactions with protein carbonyl groups are takenfor evidence of damage by oxidation, and the na-ture of the protein modification is not explicitlydefined. Two studies have examined the ability ofA� proteins to induce oxidative modifications inother proteins (34), and A� proteins may also un-dergo oxidative damage, particularly His, Tyr, andMet side chains (15, 32, 43, 132, 139). However, it isdifficult to imagine how oxidative damage to pro-teins, especially A� proteins, would lower theaqueous solubility of A� proteins and promote fi-bril formation.

Enter Lipid Membranes

Investigations into the role of lipids in AD are ex-perimentally challenging because they are chemi-cally diverse, with thousands of distinct molecularspecies present in every cell. They are also physi-cally diverse, rarely existing as monomeric speciesin solution unless protein bound. In vitro, the vastmajority form micelles or vesicles, and the lattermay be unilamellar or multilamellar. Thus lipidsuspensions generally have two or more phasesand complex interphase equilibria. This physicalheterogeneity complicates most types of physico-chemical analysis, even when pure synthetic lipidpreparations are used, and it makes some othersoutright impossible. It also impedes the measure-ment of fibril formation. Turbidity measurements,for example, are confounded by ambiguity overwhether the species causing the turbidity is proteinor lipid. Lipids markedly increase the fluorescenceof the thioflavin T apart from fibril formation,whereas assays based on Congo Red absorbanceratios entail practical restrictions on protein andlipid concentrations that limit the flexibility of thisassay. The presence of membranes can also com-plicate sample preparation, and they yield artifactswhen the halogenated solvents used to disaggre-gate A� proteins are not completely removed (56,310).

Due in part to these challenges, the evidencethat cell membranes are involved in the pathogen-esis of AD remains largely circumstantial, eventhough the amount of evidence pointing to some

type of link is overwhelming. A� proteins are pro-duced by cleavage of APP at two sites, one sitebeing located approximately at the midpoint of thetransmembrane segment (FIGURE 1). As a conse-quence, the COOH-terminal residues of A� pro-teins that were part of the APP transmembranesegment are uniformly hydrophobic. Followingtheir cleavage from APP, some investigators haveobserved that A� proteins remain associated withdetergent-resistant lipid membrane domains inthe brain (181), or with membrane-anchored APP(189). It has also been hypothesized that A� pro-teins bind to the transmembrane helices of mem-brane proteins and cause their dysfunction (199).Ultrastructural studies suggest that amyloid fibrilformation tends to occur first in portions of diffuseamyloid deposits that are closest to membranes(234, 319, 346). A�40 with the E22Q mutation (re-sponsible for hereditary cerebral hemorrhage withamyloidosis-Dutch type) will fibrillize on the sur-face membrane of human cerebrovascular smoothmuscle cells (330).

Despite a substantial hydrophobic segment, thegeneral conclusion reached by most investigatorsis that A� proteins have little affinity for neutrallipid membranes (205). Techniques such as thehydration of a mixed protein-lipid film must beused to induce the penetration of A� proteins intoa neutral lipid bilayer (83). One laboratory investi-gating A�40 and another investigating A�42 havedocumented that the proteins situate differently inmembranes depending on whether they are em-bedded in a bilayer membrane or allowed to asso-ciate with the surface of a preformed membrane(33, 106, 176). In general, anionic lipids tend toinduce A� proteins to adopt extended � structure(35, 63, 66, 76, 130, 167–169, 204, 213, 214, 313, 314,344, 352). Possible reasons for the inducement of �

structure and fibril formation by protein-lipid in-teraction have been reviewed, including the abilityof such interactions to serve as templates for struc-tural change, to increase the local concentration ofprotein, and to orient protein monomers relative toeach other (2, 113). A� proteins adopt �-helicalstructure in association with lipid membranes atlow lipid-to-protein ratios (315), at high cholesterolconcentrations (145), or in conjunction with metalions (21, 76, 77). Some investigators have found thatdetergent micelles promote �-helical structure (283),whereas others find that it promotes the formation ofoligomers with � structure (261). Spontaneous inser-tion into various membranes has been observed forA� segments (82) and for A� proteins at relatively lowmembrane surface pressures (94).

The relative abundance of various lipid classes inmembranes is altered in AD (121, 227, 247, 252,350), and altering membrane composition protectsPC12 cells from toxic effects of A� proteins (338).

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org56

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 5: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

Altered physical properties, presumably arisingfrom compositional differences, have been ob-served in hippocampal membranes of AD brains(364). Plasmalogen deficiency is frequently associ-ated with AD (102, 110 –112, 119, 120, 229). In ad-dition to having an effect on membrane physicalproperties, plasmalogens are particularly suscepti-ble to oxidative damage (156 –158). This suscepti-bility may confer on plasmalogens the ability toprotect other lipid species by diverting and trap-ping oxidizing agents (36, 117, 118, 170, 195, 196,222, 230, 242, 264, 362). Cerebral white matter isenriched in plasmalogens for unknown reasons(101).

Human epidemiological studies support a linkbetween AD and the consumption of �-3 polyun-saturated fatty acyl (PUFA) chains (103, 141, 223,224, 237, 250, 275, 323), and this association issupported by animal model and cell culture stud-ies (53–55, 126, 146, 185, 193, 350). The most prev-alent �-3 PUFA in the brain is docosahexaenoicacid (DHA), and dietary DHA supplementation al-leviates both AD-like histopathology and cognitiveimpairment in animal models (53, 55, 126, 185).The metabolism of DHA in normal brain is remark-able in several respects (159), and it is difficult toinduce a measurable deficiency of DHA-containinglipids in the brain tissue of animal models throughdietary restriction (84). Animal studies have shownthat the brain responds to a dietary deficiency ofDHA by elongating arachidonic acid (ARA) chains.Because there are no enzymes capable of desatu-rating the distal end of these PUFA chains, theresult is a marked increase in docosapentaenoicacid (DPA) chains (138).

A� proteins appear to have special relationshipsor interactions with specific lipid species. For ex-ample, the ganglioside GM1 is bound together withA� proteins in diffuse amyloid plaques (348). GM1-containing membranes promote the formation of� structure (212, 215), � structure (206), or fibrils invitro (67– 69, 115, 127, 148 –151, 165, 207, 218, 239,240, 334, 335). In several of these reports, GM1 ispresented in raft-like membrane subdomains inwhich cholesterol is a significant component, andrafts have also been discussed as an influence onthe processing of APP into A� proteins (74). Apartfrom rafts, cholesterol has been epidemiologically(289, 311) and experimentally (263, 300) linked tothe incidence of AD, and it may influence the pro-duction (108, 361), location (77, 87, 145), behavior(79, 214, 352), or toxicity (38, 78, 137, 308, 338) ofA� proteins. In models of Niemann-Pick Type Cdisease, A� proteins accumulate along with cho-lesterol (347).

Cholesterol depletion reduced the �-cleavage ofAPP and the production of A� proteins in a study ofneurons in the rat hippocampus (288). Treatment

with lovastatin/mevalonate alone, however, was in-sufficient to induce a significant effect; treatmentwith �-cyclodextrin to achieve a 70% reduction ofcellular cholesterol content was also required. It hasbeen suggested that the dependence of A� proteinproduction on cholesterol is due to the selective ac-tivity of �-secretase activity in cholesterol-dependentraft-like subdomains of the plasma membrane,whereas �-secretase activity appears to predominatein non-raft domains (95). Some �-secretase stimula-tion has also been attributed to neutral glycosphin-golipids and anionic phospholipids (152). Theefficacy of a �-secretase inhibitor has been increasedby linking it to cholesterol and thereby targeting it tomembranes (260).

The Membrane as Villain

The lipids in cell membranes are often regarded asbeing chemically unreactive and merely a physicalbarrier or a support matrix for proteins. That viewis misleading on many levels, of course, but par-ticularly so when the membrane lipids containPUFA chains and are subjected to oxidative stress.Reviews of the role of membranes in AD pathogen-esis frequently overlook the effects of oxidativestress and chemical modification of PUFA chainson membrane properties. A� proteins have aprooxidant activity toward polyunsaturated lipidsthat can be neutralized by lipophilic antioxidants,chelation of metal ions, anaerobic conditions, mu-tation of His13 or His14 to Ala, or modification ofthe Met35 side chain (232). Lipid oxidation prod-ucts and the susceptibility of lipids to oxidativedamage are both increased in AD (78, 104, 210,309). Several reports have outlined the potential fora complex interplay between cholesterol oxidationproducts and A� proteins (38, 116, 235, 241, 301,329, 356, 360).

The myriad mechanisms of oxidative stress yielddiverse chemically reactive products from PUFAchains including hydroxy- and hydroperoxy-lipidsthat undergo spontaneous decomposition, lipidfree radicals that may participate in free-radicalchain reactions, as well as fragments such as ma-londialdehyde, acrolein, and hydroxynonenal withthe potential to form adducts with proteins andnucleic acids (61, 270, 274, 293, 295, 296). Thesematerials are direct toxic threats to the tissues inwhich they are generated. Brain is the most lipid-rich organ in the body, and it contains more lipidsbearing PUFA chains than any other organ. There-fore, brain tissue is at particularly high risk forchemical injury due to highly reactive lipid oxida-tion products.

Evidence that lipid peroxidation may be involvedin the pathogenesis of AD has been extensivelyreviewed (13, 42, 46, 201). Lipid hydroperoxides

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 57

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 6: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

undergo spontaneous (nonenzymatic) decomposi-tion, and �-6 PUFA chains yield reactive �,�-un-saturated aldehydes such as 4-oxo-2(E)-nonenal(180) and 4-hydroxy-2(E)-nonenal (HNE) (97, 98),as well as eicosanoids such as isoprostanes (225,226). The prooxidant activity of A� proteins men-tioned above can catalyze the formation of HNEfrom �-6 PUFAs in the presence of copper ions(232). Isoprostanes are relatively unreactive com-pounds that have been used as biomarkers of oxi-dative stress (179, 221, 244, 268). Some reportssuggest that isoprostanes are specifically elevatedin AD (253, 255, 256), although these findings havenot been confirmed by all investigators (221, 357).Analogous products derived from �-3 PUFA chainshave been dubbed “neuroprostanes” and also havebeen put forward as indexes of oxidative stress inthe brain (14, 266, 267, 298).

Compared with isoprostanes, HNE is chemicallyreactive, with a well known propensity to formadducts with the side chains of various amino acidresidues (37, 98, 328). For this reason, HNE-proteinadducts have also been used as biomarkers of ox-idative stress (325). HNE concentrations in humanventricular fluid are �15 �M and are elevated inAD (190, 203, 210). HNE modification is also knownto inhibit proteasome function (286) and gluta-mate transport in synaptosomes (177). Togetherwith the observation that immunoreactivity of an-tibodies to HNE-modified His residues localizes toamyloid plaques (7, 294), these observations sug-gest that A� proteins not only promote lipid oxi-dation but that there may also be a mechanisticlink between the lipid oxidation products formedduring oxidative stress and A� misfolding (161).

A� proteins increase HNE production from PUFAchains in vitro that, in turn, causes covalent modifi-cation of the three His residues in A� proteins andfibril formation (231). These modifications promotethe aggregation of A� proteins into fibrils (FIGURE 2)(165, 231, 232). Moreover, they increase the ability ofA�42 to seed fibril formation by unmodified A�40(166). The effects of HNE on A� fibril formation de-pend on the size or hydrophobicity of the modifica-tion because the corresponding analog from �-3PUFA chains, 4-hydroxy-2(E)-hexenal, does not havethis effect (187). The addition of an octanoyl group tospecific Lys residues of A� proteins also appears tohave a pro-amyloidogenic effect (258).

In vivo, HNE-His epitopes are concentrated inthe vicinity of amyloid plaques, but they do notprecisely co-localize with the plaques (FIGURE 3).One possible explanation for this distribution isthat the presence of HNE-His adducts and Cu-Hiscomplexes in the same protein molecule are mu-tually exclusive. Presumably, the formation of Cu-His complexes precedes the generation of HNEand HNE-His adducts, if indeed they are responsi-

ble for generating HNE from �-6 PUFAs. Conse-quently, HNE-His epitopes would develop aroundpreexisting amyloid plaques, not within them. Asecond possibility is that A�-specific antibodyepitopes are masked by HNE modification, as ob-served in vitro for 4G8 and 6E10 epitopes (231). Athird possibility is that A� fibrils induced to formby HNE modification have relatively sparse HNE-His epitopes (as suggested by FIGURE 2), yieldingsparse fluorescence among fibrils. The fluores-cence of HNE-11S seen in FIGURE 3 may be toHNE produced in the plaque but bound to HNE-His adducts in proteins other than A� proteins.

Some investigators have focused on the role ofacrolein in the pathogenesis of AD. Acrolein is a wellknown environmental toxin and a recently recog-nized product of lipid peroxidation (324, 326, 327). Itreacts spontaneously with Lys residue side chains,forming a cyclic Nε-(3-formyl-3,4-dehydropiper-idino) derivative that has been considered abiomarker for measuring oxidative stress in AD(52). Acrolein is elevated in AD brain, it appearsto be more neurotoxic than HNE, it causes ele-vated intracellular calcium levels (192, 342), andit appears to inactivate flippase, inducing abreakdown of lipid membrane asymmetry andapoptosis (1, 59).

The Membrane as Victim

Although the foregoing discussion suggests that ADmay be caused by membrane-derived neurotoxicagents, a contrasting view is that AD is due to

FIGURE 2. Electron micrograph of fibrils inducedby HNE modificationA�42 was induced to fibrillize with HNE, then treatedwith mouse anti-His-HNE, and gold-tagged anti-mouseantibody. The focus in this image is on the gold parti-cles, but various fibril morphologies are evident, and thegold particles making anti-HNE-His antibodies are pref-erentially associated with long, relatively straight fibils.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org58

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 7: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

physical attacks on the barrier function of mem-branes (265). For example, A� proteins or theirfragments have been observed to penetrate themembrane surface (82, 94), disrupt raft-like do-mains (67), cross the membrane (341), induce iso-tropic phases (176), increase the permeability ofmembranes to various materials (154, 344), causephysical thinning (81), decrease the mobility ofhydrophobic membrane probes (169), activatephospholipase A2 (182), and promote membranefusion (81, 249). Membrane binding by A� proteinsappears to mediate some forms of neurotoxicity(21, 70, 316). Plasma membranes isolated from hu-man brain accelerate A� fibrillogenesis (339),whereas fibrillizing A� proteins disrupt the struc-ture of membranes formed from both syntheticlipids (168, 169) and whole-brain lipid extracts(353). The action of A� proteins on membranes hasbeen compared with the action of antimicrobialpeptides, and homogenates of brain with AD haveelevated antimicrobial activity (299). Among theeffects of A� proteins on membranes, however, theformation of ion channels has received the mostattention (10 –12, 93, 147, 186, 251).

A� proteins share an ability to induce channel-likeactivity in membranes with a wide variety of otheramyloid-forming proteins (259). In the case of A�, thechannels exhibit only modest cation selectivity butvery long lifetimes. A� cation channels can beblocked by Zn2�, suggesting the possibility that theymay be therapeutic targets (9), and small moleculeinhibitors of the A� calcium channel have been de-scribed (88). Ring-like structures possibly represent-ing these pores or channels have been observed inmembranes by AFM (173, 186, 259) and EM (174,175). Although such features are not always observedwith A� proteins (114, 341), there are many reportssuggesting that they are formed by other amyloido-genic proteins such as the insulin-associated poly-peptide, �-synuclein, and serum amyloid A protein(259). It has been suggested that the channel-forming

toxic properties of A� proteins on a membrane de-pends on the extent to which it has aggregated intooligomers and that this extent is concentration de-pendent (276). Model structures of the purportedchannels have been patterned after �-barrel pore-forming toxins (8, 93). This comparison is suggestednot only by the EM/AFM images but by the reactivityof A� and �-hemolysin with so-called “conforma-tion-specific” antibodies (354). Recent studies con-firming the formation of zinc-sensitive ion channelshave also exposed the potential for artifact due toresidual amounts of halogenated solvent in the A�

protein samples (56).It may be significant that truncated A� peptides

that are unable to form fibrils can neverthelessinduce ion permeability (142) and that membranesformed from highly compressible lipids are mostsensitive to A�-induced changes in permeability(297). It has been suggested that channel openingsare merely protein-induced defects in the bilayerstructure or organization that heal after a time,giving the appearance of a transient channel open-ing (114). This mechanism would be consistentwith oligomer-induced membrane thinning (297)and would help resolve the paradoxical appear-ance of many simultaneous pore structures inmembranes that exhibit only single channel con-ductances (96).

In living cells, as opposed to synthetic systems,one must consider the possibility that A� may pro-foundly affect the behavior of naturally occurringchannels and transporters. For example, A�42 in-creases HNE-modification of a glutamate trans-porter in synaptosomes (177). A� oligomers havebeen implicated in causing a pathological calciuminflux through hippocampal NMDA receptors, fol-lowed by the activation of calpain and the degra-dation of dynamic 1-a GTPase involved in synapticvesicle recycling (155). A� proteins have also beenshown to depress the surface concentration ofAMPA receptors involved in calcium regulation at

FIGURE 3. Immunofluorescent staining of the posterior parietal association area in a transgenic mouse model of Alzheimer’sdisease that expressed amyloid precursor protein with the “Swedish” K670N/M671L mutation, and presenilin 1 with theM146L mutationA: 2H4 antibodies (Covance) specific for the amino terminus A� proteins (green). B: HNE 11S specific for HNE-His adducts (red). C: A and Bsuperimposed. Possible reasons for the close association of HNE-His epitopes and A� proteins, but not precise colocalization, are discussedin the text.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 59

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 8: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

the synapse (133, 188). The dysregulation of cal-cium has long been associated with AD pathogen-esis (172) and is thought to be involved with theaccumulation of amyloid deposits (85, 208, 209)and the hyperphosphorylation of tau (25). Calciumalso promotes the conversion of oligomeric A� tofibrils (140). An extensive literature exists on therole of mitochondria in AD pathogenesis, and theaccumulation of calcium by mitochondria fol-lowed by damage to the mitochondrial membranestructure is a prominent theme in this literature(131, 246, 262).

The Lipids Associated WithApolipoprotein E

The lipids in nascent lipoprotein particles are ar-ranged in a membrane-like bilayer, surrounded byapolipoprotein molecules in a configuration oftencalled the “belt” model (FIGURE 4) (280). Apolipo-protein E (ApoE) is a focus of considerable interestbecause its isoforms are strongly associated withthe risk of developing AD. In plasma, ApoE is foundwith other proteins in lipoprotein particles as di-verse as chylomicrons and high-density lipopro-teins. In the brain and cerebrospinal fluid, ApoE isthe most abundant apolipoprotein and is found inparticles that resemble high-density lipoproteinsin both density and size (269). Among the threecommon isoforms, ApoE3 is the most common(77% of the alleles) and is therefore considered tobe the wild type. ApoE2 has an R158C substitution,whereas ApoE4 has a C112R substitution, and bothare associated with forms of hyperlipidemia (197).One copy of the ApoE4 gene confers a threefoldincreased risk of Alzheimer’s disease, whereas two

copies confer an eightfold increased risk. One copyof the ApoE2 gene, however, reduces the risk by60% (73).

In addition to the epidemiological evidence,there is an abundance of experimental evidencelinking ApoE to the pathogenesis of Alzheimer’sdisease. For example, the ApoE4 allele is associatedwith increased A� deposits in the brain, anda distinct neuropathological phenotype (278),whereas a lack of ApoE reduces A� deposition inmice (19). Amyloid plaques bind anti-ApoE anti-bodies, suggesting that ApoE is present in theseplaques (3). Indeed, ApoE copurifies with A� fromamyloid plaques (26) and may exist as a boundcomplex with A� proteins (245, 349). Some havesuggested that ApoE4 accelerates fibril formationby A�40 (18, 194, 271, 343), but its behavior in thisregard may depend on whether it is monomeric ordimeric. Others have suggested that dimeric formsinhibit fibril formation and that ApoE3 is muchmore effective at doing this because a large fractioncirculates as a disulfide-linked dimer, whereasApoE4 cannot form disulfide bonded dimers due toits C112R substitution (99, 340). Despite all of theseobservations, the reason that different isoforms ofApoE have different levels of risk for AD is not known.There are no isoform-dependent differences in theapparent structure of lipoprotein particles (280). Acase for isoform-specific direct interactions betweenApoE and A� proteins has been made (233), but clearconclusions are elusive due to problems with proteinpurity, aggregation and denaturation of the ApoEprotein, and indirect assay methods (4, 171, 198, 245,303, 307).

Others have suggested that risk of AD with ApoEisoforms may be related to differences in antioxi-dant activity (178, 217, 312). ApoE4 has no Cysresidues and, hence, no free thiol groups; ApoE3has one Cys, whereas ApoE2 has two. Free thiolgroups have significant antioxidant activity viamechanisms that differ from those of glutathione(109, 333, 359). In apoA-I, variants with a free Cysresidue side chain exhibit significantly greater an-tioxidant activity than variants without a free Cysresidue (29). Therefore, it is intriguing to speculatethat ApoE4 confers increased risk of AD, whereasApoE2 confers decreased risk, because Cys residueside chains protect lipids in lipoprotein E particlesagainst oxidative stress.

Toward a Comprehensive Theoryof AD Pathogenesis

The cause of AD is known in cases of familialdisease, but that knowledge has not clarified howsporadic disease develops, nor has it so far yieldedan effective therapy for either form of the disease.Clearly, we need a more detailed theory of AD

FIGURE 4. Nascent lipoprotein E particles consist of�-helical “belts” around the perimeter of a lipid bilayerdiskFor this arrangement and the number of protein and lipid mole-cules in a particle, there is excess protein, which may situateacross the face of the disk. When the protein is apoE3, there isone thiol group for every 45 lipids, concentrating a large antioxi-dant capacity in close proximity to the lipids.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org60

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 9: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

pathogenesis, and it seems likely that lipid mem-branes will have a prominent role in any suchtheory. As summarized above, the interactions be-tween lipid membranes and A� proteins are bi-directional: lipids damage A� proteins, and A�

proteins damage lipid membranes. These pro-cesses can operate in tandem to accelerate fibrilformation in human brain lipid extracts (231). Evenif fibril formation is only an inert by-product of thedisease-causing process, it is nonetheless a patho-gnomonic finding of AD. Therefore, it is vital tounderstand how fibrils form.

As illustrated in FIGURE 5, the interaction of lipidmembranes may constitute an amplification sys-tem for fibril formation. A� proteins in vitro onlyform fibrils at micromolar concentrations and afterdays of incubation, whereas membranes contain-ing PUFA-chains lower the protein concentrationrequirement by three orders of magnitude andshorten the time required to minutes (165, 166).The same system may also amplify multiple mech-anisms of neurotoxicity that operate indepen-dently of fibril formation and link many of theseemingly unrelated observations reviewed above.For example, inflammation due to trauma or otherfactors will promote oxidative stress and the pro-duction of reactive oxygen species that exert directneurotoxicity or lipid damage and A� protein ag-gregation. Lipid oxidation products may exertdirect toxicity. ApoE4 proteins are deficient inthiol-mediated antioxidant activity; this deficiencywould allow excess oxidative damage to the lipidsin lipoprotein particles that likewise promote A�

protein aggregation. Aging is associated with re-duced tissue antioxidant levels, accumulated oxi-dative lipid damage, and low levels of adventitialA� protein aggregation, with each process promot-ing further A� protein aggregation. Various aggre-gated forms of A� protein may form pores,channels, or other neurotoxic structures in neuro-nal membranes.

It is not yet clear whether all cases of AD arisethrough the operation of one primary pathogenicmechanism that diverges only at a late stage intovarious subtypes or whether disparate mecha-nisms operate from the beginning. For example,neurofibrillary tangles of tau protein (20, 322) oc-cur in only 70 – 80% of cases of AD (122), suggestingthat tangles reflect a response to some but not allpathogenic paths. In any case, one or more addi-tional factors must be incorporated into the amy-loid hypothesis to explain the pathogenesis of AD,and confronting the experimental challenges pre-sented by lipids and membranes may be necessaryto identify such factors. �

No conflicts of interest, financial or otherwise, are de-clared by the author(s).

The authors are supported by grants from the NationalInstitute on Aging, the Alzheimer’s Association, and theAmerican Health Assistance Foundation.

References1. Abdul HM, Butterfield DA. Protection against amyloid beta-

peptide (1-42)-induced loss of phospholipid asymmetry insynaptosomal membranes by tricyclodecan-9-xanthogenate(D609) and ferulic acid ethyl ester: implications for Alzhei-mer’s disease. BBA Mol Basis Dis 1741: 140–148, 2005.

2. Aisenbrey C, Borowik T, Bystrom R, Bokvist M, Lindstrom F,Misiak H, Sani MA, Grobner G. How is protein aggregation inamyloidogenic diseases modulated by biological mem-branes? Eur Biophys J Biophys Lett 37: 247–255, 2008.

3. Aizawa Y, Fukatsu R, Takamaru Y, Tsuzuki K, Chiba H, Ko-bayashi K, Fujii N, Takahata N. Amino-terminus truncatedapolipoprotein E is the major species in amyloid deposits inAlzheimer’s disease affected brains: a possible role for apo-lipoprotein E in Alzheimer’s disease. Brain Res 768: 208–214,1997.

4. Aleshkov S, Abraham CR, Zannis VI. Interaction of nascentApoE2, ApoE3, and ApoE4 isoforms expressed in mamma-lian cells with amyloid peptide beta (1–40). Relevance toAlzheimer’s disease. Biochemistry 36: 10571–10580, 1997.

5. Ali FE, Separovic F, Barrow CJ, Cherny RA, Fraser F, Bush AI,Masters CL, Barnham KJ. Methionine regulates copper/hy-drogen peroxide oxidation products of A beta. J Pept Sci 11:353–360, 2005.

6. Ali FE, Separovic F, Barrow CJ, Yao SG, Barnham KJ. Copperand zinc mediated oligomerisation of A beta peptides. Int JPept Res Ther 12: 153–164, 2006.

7. Ando Y, Brannstrom T, Uchida K, Nyhlin N, Nasman B, SuhrO, Yamashita T, Olsson T, El Salhy M, Uchino M, Ando M.Histochemical detection of 4-hydroxynonenal protein in Alz-heimer amyloid. J Neurol Sci 156: 172–176, 1998.

8. Arispe N. Architecture of the Alzheimer’s A beta P ion chan-nel pore. J Membrane Biol 197: 33–48, 2004.

FIGURE 5. Membrane-mediated amplification of fibrillogenesis, illustratingpossible relationships between meachanisms involved in A� proteinaggregation and neurotoxic mechanisms involved in the pathogenesis ofAlzheimer’s diseaseLipid oxidation products modify the three His residues of A�42, increasing its membraneaffinity and accelerating the conversion of A�40 into oligomers and fibrils. Oligomeric A�proteins bind copper ions while they undergo redox cycling. Highly reactive oxygen spe-cies may be generated by electrons from copper or as by-products of inflammation, in-cluding inflammation induced by trauma. ApoE4 alleles lack thiol-mediated antioxidantactivity and may allow excess oxidative damage to the lipids in lipoprotein particles. Agingby itself is associated with reduced tissue antioxidant levels and accumulated oxidativelipid damage. During any or all of these processes, direct neurotoxins may be produced,and A� proteins may form pores, channels, or other disruptive neurotoxic structures inneuronal membranes.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 61

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 10: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

9. Arispe N, Diaz JC, Simakova OA. � Ion channels.Prospects for treating Alzheimer’s disease withA� channel blockers. BBA Biomembranes 1768:1952–1965, 2007.

10. Arispe N, Pollard HB, Rojas E. Giant multilevelcation channels formed by Alzheimer-diseaseamyloid beta-protein [A-Beta-P-(1-40)] in bilayer-membranes. Proc Natl Acad Sci USA 90: 10573–10577, 1993.

11. Arispe N, Pollard HB, Rojas E. Zn2� interactionwith Alzheimer amyloid beta protein calciumchannels. Proc Natl Acad Sci USA 93: 1710–1715,1996.

12. Arispe N, Rojas E, Pollard HB. Alzheimer-diseaseamyloid beta-protein forms calcium channels inbilayer-membranes: blockade by tromethamineand aluminum. Proc Natl Acad Sci USA 90: 567–571, 1993.

13. Arlt S, Beisiegel U, Kontush A. Lipid peroxidationin neurodegeneration: new insights into Alzhei-mer’s disease. Curr Opin Lipid 13: 289–294,2002.

14. Arneson KO, Roberts LJ. Measurement of prod-ucts of docosahexaenoic acid peroxidation, neu-roprostanes, and neurofurans. LipidomicsBioactive Lipids 433: 127–143, 2007.

15. Atwood CS, Huang XD, Khatri A, Scarpa RC, KimYS, Moir RD, Tanzi RE, Roher AE, Bush AI. Cop-per catalyzed oxidation of alzheimer A beta. CellMol Biol 46: 777–783, 2000.

16. Atwood CS, Obrenovich ME, Liu TB, Chan H,Perry G, Smith MA, Martins RN. Amyloid-beta: achameleon walking in two worlds: a review of thetrophic and toxic properties of amyloid-beta.Brain Res Rev 43: 1–16, 2003.

17. Atwood CS, Perry G, Zeng H, Kato Y, Jones WD,Ling KQ, Huang XD, Moir RD, Wang DD, SayreLM, Smith MA, Chen SG, Bush AI. Copper medi-ates dityrosine cross-linking of Alzheimer’s amy-loid-beta. Biochemistry 43: 560–568, 2004.

18. Bales KR, Verina T, Cummins DJ, Du YS, DodelTC, Saura J, Fishman CE, DeLong CA, Piccardo P,Petegnief V, Ghetti B, Paul SM. Apolipoprotein Eis essential for amyloid deposition in theAPP(V717F) transgenic mouse model of Alzhei-mer’s disease. Proc Natl Acad Sci USA 96:15233–15238, 1999.

19. Bales KR, Verina T, Dodel RC, Du YS, Altstiel L,Bender M, Hyslop P, Johnstone EM, Little SP,Cummins DJ, Piccardo P, Ghetti B, Paul SM. Lackof apolipoprotein E dramatically reduces amyloidbeta- peptide deposition. Nat Gen 17: 263–264,1997.

20. Ballatore C, Lee VMY, Trojanowski JQ. Tau-me-diated neurodegeneration in Alzheimer’s diseaseand related disorders. Nat Rev Neurosci 8: 663–672, 2007.

21. Barnham KJ, Ciccotosto GD, Tickler AK, Ali FE,Smith DG, Williamson NA, Lam YH, Carrington D,Tew D, Kocak G, Volitakis I, Separovic F, BarrowCJ, Wade JD, Masters CL, Cherny RA, CurtainCC, Bush AI, Cappai R. Neurotoxic, redox-com-petent Alzheimer’s beta-amyloid is released fromlipid membrane by methionine oxidation. J BiolChem 278: 42959–42965, 2003.

22. Barnham KJ, Haeffner F, Ciccotosto GD, CurtainCC, Tew D, Beyreuther K, Carrington D, MastersCL, Cherny RA, Cappai R, Bush AI. Tyrosinegated electron transfer is key to the toxic mech-anism of Alzheimer’s disease �-amyloid. FASEB J18: 1427–1429, 2004.

23. Baruch-Suchodolsky R, Fischer B. Soluble amy-loid beta(1-28)-copper(I)/copper(II)/iron(II) com-plexes are potent antioxidants in cell-freesystems. Biochemistry 47: 7796–7806, 2008.

24. Baruch-Suchodolsky R, Fischer B. A�40, eithersoluble or aggregated, is a remarkably potentantioxidant in cell-free oxidative systems. Bio-chemistry 48: 4354–4370, 2009.

25. Baudier J, Cole RD. Phosphorylation of tau-pro-teins to a state like that in Alzheimers brain iscatalyzed by a calcium calmodulin-dependent ki-nase and modulated by phospholipids. J BiolChem 262: 17577–17583, 1987.

26. Baumann MH, Kallijarvi J, Lankinen H, Soto C,Haltia M. Apolipoprotein E includes a bindingsite which is recognized by several amyloido-genic polypeptides. Biochem J 349: 77–84, 2000.

27. Behl C, Davis JB, Lesley R, Schubert D. Hydrogenperoxide mediates amyloid beta protein toxicity.Cell 77: 817–827, 1994.

28. Berthon G. Does human beta A4 exert a protec-tive function against oxidative stress in Alzhei-mer’s disease? Med Hypotheses 54: 672–677,2000.

29. Bielicki JK, Oda MN. Apolipoprotein A-I-Milanoand apolipoprotein A-I-Paris exhibit an antioxi-dant activity distinct from that of wild-type apo-lipoprotein A-I. Biochemistry 41: 2089–2096,2002.

30. Bieschke J, Zhang Q, Powers ET, Lerner RA, KellyJW. Oxidative metabolites accelerate Alzhei-mer’s amyloidogenesis by a two-step mecha-nism, eliminating the requirement for nucleation.Biochemistry 44: 4977–4983, 2005.

31. Bishop GM, Robinson SR. The amyloid paradox:amyloid-beta-metal complexes can be neuro-toxic and neuroprotective. Brain Pathol 14: 448–452, 2004.

32. Bitan G, Tarus B, Vollers SS, Lashuel HA, Con-dron MM, Straub JE, Teplow DB. A molecularswitch in amyloid assembly: Met(35) and amyloidbeta-protein oligomerization. J Am Chem Soc125: 15359–15365, 2003.

33. Bokvist M, Lindstrom F, Watts A, Grobner G.Two types of Alzheimer’s beta-amyloid (1-40)peptide membrane interactions: aggregationpreventing transmembrane anchoring versus ac-celerated surface fibril formation. J Mol Biol 335:1039–1049, 2004.

34. Boyd-Kimball D, Castegna A, Sultana R, Poon HF,Petroze R, Lynn BC, Klein JB, Butterfield DA.Proteomic identification of proteins oxidized byA beta(1-42) in synaptosomes: implications forAlzheimer’s disease. Brain Res 1044: 206–215,2005.

35. Brezesinski G, Maltseva E, Mohwald H. Adsorp-tion of amyloid beta (1-40) peptide at liquid in-terfaces. Z Phys Chem 221: 95–111, 2007.

36. Brosche T, Platt D. Mini-review: The biologicalsignificance of plasmalogens in defense againstoxidative damage. Exp Gerontol 33: 363–369,1998.

37. Bruenner BA, Jones AD, German JB. Direct char-acterization of protein adducts of the lipid-per-oxidation product 4-hydroxy-2-nonenal usingelectrospray mass-spectrometry. Chem Res Tox8: 552–559, 1995.

38. Bryleva EY, Rogers MA, Chang CCY, Buen F,Harris BT, Rousselet E, Seidah NG, Oddo S,LaFerla FM, Spencer TA, Hickey WF, Chang TY.ACAT1 gene ablation increases 24(S)-hydroxy-cholesterol content in the brain and amelioratesamyloid pathology in mice with AD. Proc NatlAcad Sci USA 107: 3081–3086, 2010.

39. Bush AI. The metallobiology of Alzheimer’s dis-ease. Trends Neurosci 26: 207–214, 2003.

40. Butterfield DA. Amyloid beta-peptide (1-42)-in-duced oxidative stress and neurotoxicity: impli-cations for neurodegeneration in Alzheimer’sdisease brain. A review. Free Radic Res 36: 1307–1313, 2002.

41. Butterfield DA. Proteomics: a new approach toinvestigate oxidative stress in Alzheimer’s dis-ease brain. Brain Res 1000: 1–7, 2004.

42. Butterfield DA, Boyd-Kimball D. Amyloid beta-peptide(1-42) contributes to the oxidative stressand neurodegeneration found in Alzheimer dis-ease brain. Brain Pathol 14: 426–432, 2004.

43. Butterfield DA, Boyd-Kimball D. The critical roleof methionine 35 in Alzheimer’s amyloid-beta-peptide (1-42)-induced oxidative stress and neu-rotoxicity. Biochim Biophys Acta 1703: 149–156,2005.

44. Butterfield DA, Boyd-Kimball D, Castegna A. Pro-teomics in Alzheimer’s disease: insights into po-tential mechanisms of neurodegeneration. JNeurochem 86: 1313–1327, 2003.

45. Butterfield DA, Bush AI. Alzheimer’s amyloid be-ta-peptide (1-42): involvement of methionine res-idue 35 in the oxidative stress and neurotoxicityproperties of this peptide. Neurobiol Aging 25:563–568, 2004.

46. Butterfield DA, Castegna A, Lauderback CM,Drake J. Evidence that amyloid beta-peptide-in-duced lipid peroxidation and its sequelae in Alz-heimer’s disease brain contribute to neuronaldeath. Neurobiol Aging 23: 655–664, 2002.

47. Butterfield DA, Galvan V, Lange MB, Tang HD,Sowell RA, Spilman P, Fombonne J, Gorostiza O,Zhang JL, Sultana R, Bredesen DE. In vivo oxida-tive stress in brain of Alzheimer disease trans-genic mice: requirement for methionine 35 inamyloid beta-peptide of APP. Free Radic BiolMed 48: 136–144, 2010.

48. Butterfield DA, Hensley K, Harris M, Mattson MP,Carney JM. Beta-amyloid peptide free-radicalfragments initiate synaptosomal lipoperoxida-tion in a sequence-specific fashion: implicationsto Alzheimers-disease. Biochem Biophys ResCommun 200: 710–715, 1994.

49. Butterfield DA, Kanski J. Methionine residue 35 iscritical for the oxidative stress and neurotoxicproperties of Alzheimer’s amyloid beta-peptide1-42. Peptides 23: 1299–1309, 2002.

50. Butterfield DA, Lauderback CM. Lipid peroxida-tion and protein oxidation in Alzheimer’s diseasebrain: potential causes and consequences involv-ing amyloid beta-peptide-associated free radicaloxidative stress. Free Radic Biol Med 32: 1050–1060, 2002.

51. Butterfield DA, Reed T, Newman SF, Sultana R.Roles of amyloid beta-peptide-associated oxida-tive stress and brain protein modifications in thepathogenesis of Alzheimer’s disease and mildcognitive impairment. Free Radic Biol Med 43:658–677, 2007.

52. Calingasan NY, Uchida K, Gibson GE. Protein-bound acrolein: a novel marker of oxidativestress in Alzheimer’s disease. J Neurochem 72:751–756, 1999.

53. Calon F, Cole G. Neuroprotective action ofomega-3 polyunsaturated fatty acids againstneurodegenerative diseases: evidence from ani-mal studies. Prost Leuk Essen Fatty Acids 77:287–293, 2007.

54. Calon F, Lim GP, Morihara T, Yang FS, Ubeda O,Salem N, Frautschy SA, Cole GM. Dietary n-3polyunsaturated fatty acid depletion activatescaspases and decreases NMDA receptors in thebrain of a transgenic mouse model of Alzheimer’sdisease. Eur J Neurosci 22: 617–626, 2005.

55. Calon F, Lim GP, Yang FS, Morihara T, Teter B,Ubeda O, Rostaing P, Triller A, Salem N, AsheKH, Frautschy SA, Cole GM. Docosahexaenoicacid protects from dendritic pathology in an Alz-heimer’s disease mouse model. Neuron 43: 633–645, 2004.

56. Capone R, Quiroz FG, Prangkio P, Saluja I, SauerAM, Bautista MR, Turner RS, Yang J, Mayer M.Amyloid-beta-induced ion flux in artificial lipidbilayers and neuronal cells: resolving a contro-versy. Neurotox Res 16: 1–13, 2009.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org62

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 11: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

57. Castegna A, Aksenov M, Aksenova M, Thong-boonkerd V, Klein JB, Pierce WM, Booze R,Markesbery WR, Butterfield DA. Proteomic iden-tification of oxidatively modified proteins in Alz-heimer’s disease brain. Part 1: creatine kinase bb,glutamine synthase, and ubiquitin carboxy-termi-nal hydrolase L-1. Free Radic Biol Med 33: 562–571, 2002.

58. Castegna A, Aksenov M, Thongboonkerd V,Klein JB, Pierce WM, Booze R, Markesbery WR,Butterfield DA. Proteomic identification of oxida-tively modified proteins in Alzheimer’s diseasebrain. Part II: dihydropyrimidinase-related pro-tein 2, alpha-enolase and heat shock cognate 71.J Neurochem 82: 1524–1532, 2002.

59. Castegna A, Lauderback CM, Mohmmad-AbdulH, Butterfield DA. Modulation of phospholipidasymmetry in synaptosomal membranes by thelipid peroxidation products, 4-hydroxynonenaland acrolein: implications for Alzheimer’s dis-ease. Brain Res 1004: 193–197, 2004.

60. Castegna A, Thongboonkerd V, Klein JB, Lynn B,Markesbery WR, Butterfield DA. Proteomic iden-tification of nitrated proteins in Alzheimer’s dis-ease brain. J Neurochem 85: 1394–1401, 2003.

61. Castellani RJ, Perry G, Harris PLR, Cohen ML,Sayre LM, Salomon RG, Smith MA. Advancedlipid peroxidation end-products in Alexander’sdisease. Brain Res 787: 15–18, 1998.

62. Caughey B, Lansbury PT. Protofibrils, pores, fi-brils, and neurodegeneration: separating the re-sponsible protein aggregates from the innocentbystanders. Ann Rev Neurosci 26: 267–298,2003.

63. Chauhan A, Ray I, Chauhan VPS. Interaction ofamyloid beta-protein with anionic phospholipids:possible involvement of Lys(28) and C-terminusaliphatic amino acids. Neurochem Res 25: 423–429, 2000.

64. Cherny RA, Atwood CS, Xilinas ME, Gray DN,Jones WD, Mclean CA, Barnham KJ, Volitakis I,Fraser FW, Kim YS, Huang XD, Goldstein LE, MoirRD, Lim JT, Beyreuther K, Zheng H, Tanzi RE, Mas-ters CL, Bush AI. Treatment with a copper-zincchelator markedly and rapidly inhibits beta-amy-loid accumulation in Alzheimer’s disease trans-genic mice. Neuron 30: 665–676, 2001.

65. Cherny RA, Legg JT, Mclean CA, Fairlie DP,Huang XD, Atwood CS, Beyreuther K, Tanzi RE,Masters CL, Bush AI. Aqueous dissolution of Alz-heimer’s disease A beta amyloid deposits by bio-metal depletion. J Biol Chem 274: 23223–23228,1999.

66. Chi EY, Ege C, Winans A, Majewski J, Wu GH,Kjaer K, Lee KYC. Lipid membrane templates theordering and induces the fibrillogenesis of Alz-heimer’s disease amyloid-beta peptide. Proteins72: 1–24, 2008.

67. Chi EY, Frey SL, Lee KYC. Ganglioside GM1-mediated amyloid-beta fibrillogenesis and mem-brane disruption. Biochemistry 46: 1913–1924,2007.

68. Choo-Smith LP, Garzon-Rodriguez W, Glabe CG,Surewicz WK. Acceleration of amyloid fibril for-mation by specific binding of Abeta-(1-40)peptide to ganglioside-containing membranevesicles. J Biol Chem 272: 22987–22990, 1997.

69. Choo-Smith LP, Surewicz WK. The interaction be-tween Alzheimer amyloid beta(1-40) peptide andganglioside GM1-containing membranes. FEBSLett 402: 95–98, 1997.

70. Ciccotosto GD, Tew D, Curtain CC, smith D, Car-rington D, Masters CL, Bush AI, Cherny RA, Cap-pai R, Barnham KJ. Enhanced toxicity and cellularbinding of a modified amyloid beta peptide witha methionine to valine substitution. J Biol Chem279: 28425–42538, 2004.

71. Cirrito JR, Yamada KA, Finn MB, Sloviter RS,Bales KR, May PC, Schoepp DD, Paul SM, Menn-erick S, Holtzman DM. Synaptic activity regulatesinterstitial fluid amyloid-beta levels in vivo. Neu-ron 48: 913–922, 2005.

72. Cleary JP, Walsh DM, Hofmeister JJ, ShankarGM, Kuskowski MA, Selkoe DJ, Ashe KH. Naturaloligomers of the amyloid-protein specifically dis-rupt cognitive function. Nat Neurosci 8: 79–84,2005.

73. Corder EH, Saunders AM, Strittmatter WJ, Sch-mechel DE, Gaskell PC, Small GW, Roses AD,Haines JL, Pericakvance MA. Gene dose of apo-lipoprotein-E type-4 allele and the risk of Al-zheimers-disease in late-onset families. Science261: 921–923, 1993.

74. Cordy JM, Hooper NM, Turner AJ. The involve-ment of lipid rafts in Alzheimer’s disease. MolMembr Biol 23: 111–122, 2006.

75. Cuajungco MP, Goldstein LE, Nunomura A,Smith MA, Lim JT, Atwood CS, Huang X, FarragYW, Perry G, Bush AI. Evidence that the �-amy-loid plaques of Alzheimer’s disease represent theredox-silencing and entombment of abeta byzinc. J Biol Chem 275: 19439–19442, 2000.

76. Curtain CC, Ali F, Volitakis I, Cherny RA, Norton RS,Beyreuther K, Barrow CJ, Masters CL, Bush AI,Barnham KJ. Alzheimer’s eisease amyloid-betabinds copper and zinc to generate an allosteri-cally ordered membrane-penetrating structurecontaining superoxide dismutase-like subunits. JBiol Chem 276: 20466–20473, 2001.

77. Curtain CC, Ali FE, Smith DG, Bush AI, MastersCL, Barnham KJ. Metal ions, pH, and cholesterolregulate the interactions of Alzheimer’s diseaseamyloid-beta peptide with membrane lipid. J BiolChem 278: 2977–2982, 2003.

78. Cutler RG, Kelly J, Storie K, Pedersen WA, Tam-mara A, Hatanpaa K, Troncoso JC, Mattson MP.Involvement of oxidative stress-induced abnor-malities in ceramide and cholesterol metabolismin brain aging and Alzheimer’s disease. Proc NatlAcad Sci USA 101: 2070–2075, 2004.

79. D’Errico G, Vitiello G, Ortona O, Tedeschi A,Ramunno A, D’Ursi AM. Interaction between Alz-heimer’s A beta(25-35) peptide and phospholipidbilayers: the role of cholesterol. Biochim BiophysActa 1778: 2710–2716, 2008.

80. Dalle-Donne I, Rossi R, Giustarini D, Gagliano N,Lusini L, Milzani A, Di Simplicio P, Colombo R.Actin carbonylation: from a simple marker of pro-tein oxidation to relevant signs of severe func-tional impairment. Free Radic Biol Med 31: 1075–1083, 2001.

81. Dante S, Hauss T, Brandt A, Dencher NA. Mem-brane fusogenic activity of the Alzheimer’s pep-tide A beta(1-42) demonstrated by small-angleneutron scattering. J Mol Biol 376: 393–404,2008.

82. Dante S, Hauss T, Dencher NA. Insertion of ex-ternally administered amyloid beta peptide25-35 and perturbation of lipid bilayers. Bio-chemistry 42: 13667–13672, 2003.

83. de Planque MRR, Raussens V, Contera SA, RijkersDTS, Liskamp RMJ, Ruysschaert JM, Ryan JF, Sepa-rovic F, Watts A. beta-sheet structured beta-am-yloid(1-40) perturbs phosphatidylcholine modelmembranes. J Mol Biol 368: 982–997, 2007.

84. DeMar JC, Ma KZ, Bell JM, Rapoport SI. Half-lives of docosahexaenoic acid in rat brain phos-pholipids are prolonged by 15 weeks ofnutritional deprivation of n-3 polyunsaturatedfatty acids. J Neurochem 91: 1125–1137, 2004.

85. Demuro A, Parker I, Stutzmann GE. Calcium sig-naling and amyloid toxicity in Alzheimer’s dis-ease. J Biol Chem 285: 12463–12468, 2010.

86. Deshpande A, Kawai H, Metherate R, Glabe CG,Busciglio J. A role for synaptic zinc in activity-dependent A beta oligomer formation and accu-mulation at excitatory synapses. J Neurosci 29:4004–4015, 2009.

87. Devanathan S, Salamon Z, Lindblom G, GrobnerG, Tollin G. Effects of sphingomyelin, cholesteroland zinc ions on the binding, insertion and ag-gregation of the amyloid A beta(1-40) peptide insolid-supported lipid bilayers. FEBS J 273: 1389–1402, 2006.

88. Diaz JC, Simakova O, Jacobson KA, Arispe N,Pollard HB. Small molecule blockers of the Alz-heimer A beta calcium channel potently protectneurons from A beta cytotoxicity. Proc Natl AcadSci USA 106: 3348–3353, 2009.

89. Dikalov SI, Vitek MP, Maples KR, Mason RP. Am-yloid beta peptides do not form peptide-derivedfree radicals spontaneously, but can enhancemetal-catalyzed oxidation of hydroxylamines tonitroxides. J Biol Chem 274: 9392–9399, 1999.

90. Donnelly PS, Xiao ZG, Wedd AG. Copper andAlzheimer’s disease. Curr Opin Chem Biol 11:128–133, 2007.

91. Drake J, Link CD, Butterfield DA. Oxidativestress precedes fibrillar deposition of Alzhei-mer’s disease amyloid beta-peptide (1-42) in atransgenic Caenorhabditis elegans model. Neu-robiol Aging 24: 415–420, 2003.

92. Drew SC, Noble CJ, Masters CL, Hanson GR,Barnham KJ. Pleomorphic copper coordinationby Alzheimer’s disease amyloid-beta peptide. JAm Chem Soc 131: 1195–1207, 2009.

93. Durell SR, Guy HR, Arispe N, Rojas E, PollardHB. Theoretical-models of the ion-channel struc-ture of amyloid beta-protein. Biophys J 67: 2137–2145, 1994.

94. Ege C, Lee KYC. Insertion of Alzheimer’s A beta40 peptide into lipid monolayers. Biophys J 87:1732–1740, 2004.

95. Ehehalt R, Keller P, Haass C, Thiele C, Simons K.Amyloidogenic processing of the Alzheimerbeta-amyloid precursor protein depends on lipidrafts. J Cell Biol 160: 113–123, 2003.

96. Eliezer D. Amyloid ion channels: a porous argu-ment or a thin excuse? J Gen Physiol 128: 631–633, 2006.

97. Esterbauer H, Benedetti A, LANGJ, , Fulceri R,Fauler G, Comporti M. Studies on the mechanismof formation of 4-hydroxynonenal during micro-somal lipid-peroxidation. Biochim Biophys Acta876: 154–166, 1986.

98. Esterbauer H, Schaur RJ, Zollner H. Chemistryand biochemistry of 4-hydroxynonenal, malonal-dehyde and related aldehydes. Free Radic BiolMed 11: 81–128, 1991.

99. Evans KC, Berger EP, Cho CG, Weisgraber KH,Lansbury PT. Apolipoprotein E is a kinetic but nota thermodynamic inhibitor of amyloid formation:implications for the pathogenesis and treatmentof Alzheimer disease. Proc Natl Acad Sci USA 92:763–767, 1995.

100. Faller P, Hureau C. Bioinorganic chemistry ofcopper and zinc ions coordinated to amyloid-beta peptide. Dalton Trans 21: 1080–1094, 2009.

101. Farooqui AA, Horrocks LA. Plasmalogens: Work-horse lipids of membranes in normal and injuredneurons and glia. Neuroscientist 7: 232–245,2001.

102. Farooqui AA, Rapoport SI, Horrocks LA. Mem-brane phospholipid alterations in Alzheimer’sdisease: Deficiency of ethanolamine plasmalo-gens. Neurochem Res 22: 523–527, 1997.

103. Friedland RP. Fish consumption and the risk ofAlzheimer disease - Is it time to make dietaryrecommendations? Arch Neurol 60: 923–924,2003.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 63

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 12: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

104. Galbusera C, Facheris M, Magni F, Galimberti G,Sala G, Tremolada L, Isella V, Guerini FR, Appol-lonio I, Galli-Kienle M, Ferrarese C. Increasedsusceptibility to plasma lipid peroxidation in Alz-heimer disease patients. Curr Alzheimer Res 1:103–109, 2004.

105. Garzon-Rodriguez W, Yatsimirsky AK, Glabe CG.Binding of Zn(II), Cu(II), and Fe(II) ions to Alzhei-mer’s A beta peptide studied by fluorescence.Bioorg Med Chem Lett 9: 2243–2248, 1999.

106. Gehman JD, O’Brien CC, Shabanpoor F, WadeJD, Separovic F. Metal effects on the membraneinteractions of amyloid-beta peptides. Eur Bio-phys J Biophys Lett 37: 333–344, 2008.

107. Ghezzi P, Bonetto V. Redox proteomics: Identifi-cation of oxidatively, modified proteins. Pro-teomics 3: 1145–1153, 2003.

108. Ghribi O, Larsen B, Schrag M, Herman MM. Highcholesterol content in neurons increases BACE,beta-amyloid, and phosphorylated tau levels inrabbit hippocampus. Exp Neurol In Press: 2006.

109. Giles GI, Jacob C. Reactive sulfur species: anemerging concept in oxidative stress. Biol Chem383: 375–388, 2002.

110. Ginsberg L, Rafique S, Xuereb JH, Rapoport SI,Gershfeld NL. Disease and anatomic specificityof ethanolamine plasmalogen deficiency in Al-zheimers-disease brain. Brain Res 698: 223–226,1995.

111. Ginsberg L, Xuereb JH, Gershfeld NL. Mem-brane instability, plasmalogen content, and Alz-heimer’s disease. J Neurochem 70: 2533–2538,1998.

112. Goodenowe DB, Cook LL, Liu J, Lu Y, Jayas-inghe DA, Ahiahonu PWK, Heath D, YamazakiY, Flax J, Krenitsky KF, Sparks DL, Lerner A,Friedland RP, Kudo T, Kamino K, Morihara T,Takeda M, Wood PL. Peripheral ethanolamineplasmalogen deficiency: a logical causative factorin Alzheimer’s disease and dementia. J Lipid Res48: 2485–2498, 2007.

113. Gorbenko GP, Kinnunen PKJ. The role of lipid-protein interactions in amyloid-type protein fibrilformation. Chem Phys Lipids 141: 72–82, 2006.

114. Green JD, Kreplak L, Goldsbury C, Blatter XL,Stolz M, Cooper GS, Seelig A, Kist-Ler J, Aebi U.Atomic force microscopy reveals defects withinmica supported lipid bilayers induced by theamyloidogenic human amylin peptide. J Mol Biol342: 877–887, 2004.

115. Gylys KH, Fein JA, Yang F, Miller CA, Cole GM.Increased cholesterol in A beta-positive nerveterminals from Alzheimer’s disease cortex. Neu-robiol Aging 28: 8–17, 2007.

116. Haeffner F, Smith DG, Barnham KJ, Bush AI.Model studies of cholesterol and ascorbate oxi-dation by copper complexes: relevance to Alz-heimer’s disease beta-amyloid metallochemistry.J Inorg Biochem 99: 2403–2422, 2005.

117. Hahnel D, Beyer K, Engelmann B. Inhibition ofperoxyl radical-mediated lipid oxidation by plas-malogen phospholipids and alpha-tocopherol.Free Radic Biol Med 27: 1087–1094, 1999.

118. Hahnel D, Huber T, Kurze V, Beyer K, EngelmannB. Contribution of copper binding to the inhibi-tion of lipid oxidation by plasmalogen phospho-lipids. Biochem J 340: 377–383, 1999.

119. Han X. Lipid alterations in the earliest clinicallyrecognizable stage of Alzheimer’s disease: impli-cation of the role of lipids in the pathogenesis ofAlzheimer’s disease. Curr Alzheimer Res 2: 65–77, 2005.

120. Han XL, Holtzman DM, McKeel DW. Plasmalogendeficiency in early Alzheimer’s disease subjectsand in animal models: molecular characterizationusing electrospray ionization mass spectrometry.J Neurochem 77: 1168–1180, 2001.

121. Han XL, Holtzman DM, McKeel DW, Kelley J,Morris JC. Substantial sulfatide deficiency andceramide elevation in very early Alzheimer’s dis-ease: potential role in disease pathogenesis. JNeurochem 82: 809–818, 2002.

122. Hansen LM, Masliah E, Galasko D, Terry RD.Plaque-only Alzheimer-disease is usually theLewy body variant, and vice-versa. J NeuropathExp Neurol 52: 648–654, 1993.

123. Hardy J, Allsop D. Amyloid deposition as thecentral event in the etiology of Alzheimers-dis-ease. Trends Pharmacol Sci 12: 383–388, 1991.

124. Hardy JA, Higgins GA. Alzheimers-disease: theamyloid cascade hypothesis. Science 256: 184–185, 1992.

125. Harper JD, Wong SS, Lieber CM, Lansbury PT.Observation of metastable A beta amyloidprotofibrils by atomic force microscopy. ChemBiol 4: 119–125, 1997.

126. Hashimoto M, Tanabe Y, Fujii Y, Kikuta T, ShibataH, Shido O. Chronic administration of docosa-hexaenoic acid ameliorates the impairment ofspatial cognition learning ability in amyloid beta-infused rats. J Nutr 135: 549–555, 2005.

127. Hayashi H, Kimura N, Yamaguchi H, Hasegawa K,Yokoseki T, Shibata M, Yamamoto N, MichikawaM, Yoshikawa Y, Terao K, Matsuzaki K, LemereCA, Selkoe DJ, Naiki H, Yanagisawa K. A seed forAlzheimer amyloid in the brain. J Neurosci 24:4894–4902, 2004.

128. Hensley K, Carney JM, Mattson MP, AksenovaM, Harris M, Wu JF, Floyd RA, Butterfield DA. Amodel for beta-amyloid aggregation and neuro-toxicity based on free radical generation by thepeptide: relevance to Alzheimer disease. ProcNatl Acad Sci USA 91: 3270–3274, 1994.

129. Hensley K, Hall N, Subramaniam R, Cole P, HarrisM, Aksenov M, Aksenova M, Gabbita SP, Wu JF,Carney JM, Lovell MA, Markesbery WR, Butter-field DA. Brain regional correspondence be-tween Alzheimers-disease histopathology andbiomarkers of protein oxidation. J Neurochem65: 2146–2156, 1995.

130. Hertel C, Terzi E, Hauser N, Jakob-Rotne R,Seelig J, Kemp JA. Inhibition of the electrostaticinteraction between beta-amyloid peptide andmembranes prevents beta-amyloid-induced tox-icity. Proc Natl Acad Sci USA 94: 9412–9416,1997.

131. Hirai K, Aliev G, Nunomura A, Fujioka H, RussellRL, Atwood CS, Johnson AB, Kress Y, Vinters HV,Tabaton M, Shimohama S, Cash AD, Siedlak SL,Harris PLR, Jones PK, Petersen RB, Perry G,Smith MA. Mitochondrial abnormalities in Alzhei-mer’s disease. J Neurosci 21: 3017–3023, 2001.

132. Hou LM, Kang I, Marchant RE, Zagorski MG.Methionine 35 oxidation reduces fibril assemblyof the amyloid A beta-(1-42) peptide of Alzhei-mer’s disease. J Biol Chem 277: 40173–40176,2002.

133. Hsieh H, Boehm J, Sato C, Iwatsubo T, Tomita T,Sisodia S, Malinow R. AMPAR removal underliesA beta-induced synaptic depression and den-dritic spine loss. Neuron 52: 831–843, 2006.

134. Huang XD, Atwood CS, Hartshorn MA, MulthaupG, Goldstein LE, Scarpa RC, Cuajungco MP, GrayDN, Lim J, Moir RD, Tanzi RE, Bush AI. The Abeta peptide of Alzheimer’s disease directly pro-duces hydrogen peroxide through metal ion re-duction. Biochemistry 38: 7609–7616, 1999.

135. Huang XD, Cuajungco MP, Atwood CS, Hart-shorn MA, Tyndall JDA, Hanson GR, Stokes KC,Leopold M, Multhaup G, Goldstein LE, ScarpaRC, Saunders AJ, Lim J, Moir RD, Glabe C,Bowden EF, Masters CL, Fairlie DP, Tanzi RE,Bush AI. Cu(II) potentiation of Alzheimer A betaneurotoxicity: correlation with cell-free hydrogenperoxide production and metal reduction. J BiolChem 274: 37111–37116, 1999.

136. Hung LW, Ciccotosto GD, Giannakis E, Tew DJ,Perez K, Masters CL, Cappai R, Wade JD, Barn-ham KJ. Amyloid-beta peptide (A beta) neuro-toxicity is modulated by the rate of peptideaggregation: A beta dimers and trimers correlatewith neurotoxicity. J Neurosci 28: 11950–11958,2008.

137. Hutter-Paier B, Huttunen HJ, Puglielli L, EckmanCB, Kim DY, Hofmeister A, Moir RD, Domnitz SB,Frosch MP, Windisch M, Kovacs DM. The ACATinhibitor CP-113,818 markedly reduces amyloidpathology in a mouse model of Alzheimer’s dis-ease. Neuron 44: 227–238, 2004.

138. Igarashi M, Gao F, Kim HW, Ma KZ, Bell JM,Rapoport SI. Dietary n-6 PUFA deprivation for 15weeks reduces arachidonic acid concentrationswhile increasing n-3 PUFA concentrations in or-gans of post-weaning male rats. BBA Mol CellBiol Lipids 1791: 132–139, 2009.

139. Inoue K, Garner C, Ackermann BL, Oe T, Blair IA.Liquid chromatography/tandem mass spectrom-etry characterization of oxidized amyloid betapeptides as potential biomarkers of Alzheimer’sdisease. Rapid Commun Mass Spectrom 20: 911–918, 2006.

140. Isaacs AM, Senn DB, Yuan M, Shine JP, YanknerBA. Acceleration of amyloid beta -peptide aggre-gation by physiological concentrations of cal-cium. J Biol Chem 281: 27916–27923, 2006.

141. Issa AM, Mojica WA, Morton SC, Traina S, New-berry SJ, Hilton LG, Garland RH, MacLean CH.The efficacy of omega-3 fatty acids on cognitivefunction in aging and dementia: a systematic re-view. Dement Geriatr Cogn Disord 21: 88–96,2006.

142. Jang H, Arce FT, Ramachandran S, Capone R,Azimova R, Kagan BL, Nussinov R, Lal R. Trun-cated �-amyloid peptide channels provide an al-ternative mechanism for Alzheimer’s Disease andDown syndrome. Proc Natl Acad Sci USA 107:6838–6543, 2010.

143. Jarrett JT, Berger EP, Lansbury PT. The carboxyterminus of the beta amyloid protein is critical forthe seeding of amyloid formation: implicationsfor the pathogenesis of Alzheimer’s disease. Bio-chemistry 32: 4693–4697, 1993.

144. Jeandel C, Nicolas MB, Dubois F, Nabet-Bel-leville F, Penin F, Cuny G. Lipid peroxidation andfree radical scavengers in Alzheimer’s disease.Gerontology 35: 275–282, 1989.

145. Ji SR, Wu Y, Sui SF. Cholesterol is an importantfactor affecting the membrane insertion of beta-amyloid peptide (A beta 1-40), which may poten-tially inhibit the fibril formation. J Biol Chem 277:6273–6279, 2002.

146. Johansson AS, Garlind A, Berglind-Dehlin F,Karlsson G, Edwards K, Gellerfors P, Ekholm-Pettersson F, Palmblad J, Lannfelt L. Docosa-hexaenoic acid stabilizes soluble amyloid-betaprotofibrils and sustains amyloid-beta-inducedneurotoxicity in vitro. FEBS J 274: 990–1000,2007.

147. Kagan BL, Azimov R, Azimova R. Amyloid pep-tide channels. J Membrane Biol 202: 1–10, 2004.

148. Kakio A, Nishimoto S, Yanagisawa K, KozutsumiY, Matsuzaki K. Cholesterol-dependent forma-tion of GM1 ganglioside-bound amyloid beta-protein, an endogenous seed for Alzheimeramyloid. J Biol Chem 276: 24985–24990, 2001.

149. Kakio A, Nishimoto S, Yanagisawa K, KozutsumiY, Matsuzaki K. Interactions of amyloid beta-pro-tein with various gangliosides in raft-like mem-branes: importance of GM1 ganglioside-boundform as an endogenous seed for Alzheimer am-yloid. Biochemistry 41: 7385–7390, 2002.

150. Kakio A, Nishimoto SI, Kozutsumi Y, Matsuzaki K.Formation of a membrane-active form of amyloidbeta-protein in raft-like model membranes.Biochem Biophys Res Commun 303: 514–518,2003.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org64

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 13: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

151. Kakio A, Yano Y, Takai D, Kuroda Y, MatsumotoO, Kozutsumi Y, Matsuzaki K. Interaction be-tween amyloid beta-protein aggregates andmembranes. J Pept Sci 10: 612–621, 2004.

152. Kalvodova L, Kahya N, Schwille P, Ehehalt R,Verkade P, Drechsel D, Simons K. Lipids as mod-ulators of proteolytic activity of BACE: involve-ment of cholesterol, glycosphingolipids, andanionic phospholipids in vitro. J Biol Chem 280:36815–36823, 2005.

153. Karr JW, Szalai VA. Cu(II) binding to monomeric,oligomeric, and fibrillar forms of the Alzheimer’sdisease amyloid-beta peptide. Biochemistry 47:5006–5016, 2008.

154. Kayed R, Sokolov Y, Edmonds B, McIntire TM,Milton SC, Hall JE, Glabe CG. Permeabilization oflipid bilayers is a common conformation-depen-dent activity of soluble amyloid oligomers in pro-tein misfolding diseases. J Biol Chem 279:46363–46366, 2004.

155. Kelly BL, Ferreira A. Beta-amyloid-induced dy-namin 1 degradation is mediated by N-methyl-D-aspartate receptors in hippocampal neurons. JBiol Chem 281: 28079–28089, 2006.

156. Khaselev N, Murphy RC. Susceptibility of plasme-nyl glycerophosphoethanolamine lipids contain-ing arachidonate to oxidative degradation. FreeRadic Biol Med 26: 275–284, 1999.

157. Khaselev N, Murphy RC. Peroxidation of arachid-onate containing plasmenyl glycerophosphocho-line: facile oxidation of esterified arachidonate atcarbon-5. Free Radic Biol Med 29: 620–632,2000.

158. Khaselev N, Murphy RC. Structural characteriza-tion of oxidized phospholipid products derivedfrom arachidonate-containing plasmenyl glycero-phosphocholine. J Lipid Res 41: 564–572, 2000.

159. Kim HY. Novel metabolism of docosahexaenoicacid in neural cells. J Biol Chem 282: 18661–18665, 2007.

160. Kitazawa M, Cheng D, LaFerla FM. Chronic cop-per exposure exacerbates both amyloid and taupathology and selectively dysregulates cdk5 in amouse model of AD. J Neurochem 108: 1550–1560, 2009.

161. Komatsu H, Liu L, Murray IV, Axelsen PH. Amechanistic link between oxidative stress andmembrane mediated amyloidogenesis revealedby infrared spectroscopy. BBA Biomembranes1768: 1913–1922, 2007.

162. Kontush A. Amyloid-�: an antioxidant that be-comes a pro-oxidant and critically contributes toAlzheimer’s disease. Free Radic Biol Med 31:1120–1131, 2001.

163. Kontush A, Berndt C, Weber W, Akopyan V, ArltS, Schippling S, Beisiegel U. Amyloid-� is an an-tioxidant for lipoproteins in cerebrospinal fluidand plasma. Free Radic Biol Med 30: 119–128,2001.

164. Kontush A, Donarski N, Beisiegel U. Resistanceof human cerebrospinal fluid to in vitro oxidationis directly related to its amyloid-beta content.Free Radic Res 35: 507–517, 2001.

165. Koppaka V, Axelsen PH. Accelerated accumula-tion of amyloid beta proteins on oxidatively dam-aged lipid membranes. Biochemistry 39: 10011–10016, 2000.

166. Koppaka V, Paul C, Murray IVJ, Axelsen PH. Earlysynergy between A�42 and oxidatively damagedmembranes in promoting amyloid fibril formationby A�40. J Biol Chem 278: 36277–36284, 2003.

167. Kremer JJ, Murphy RM. Kinetics of adsorption ofbeta-amyloid peptide A beta(1-40) to lipid bilay-ers. J Biochem Biophys Methods 57: 159–169,2003.

168. Kremer JJ, Pallitto MM, Sklansky DJ, MurphyRM. Correlation of �-amyloid aggregate size andhydrophobicity with decreased bilayer fluidity ofmodel membranes. Biochemistry 39: 10309–10318, 2000.

169. Kremer JJ, Sklansky DJ, Murphy RM. Profile ofchanges in lipid bilayer structure caused by beta-amyloid peptide. Biochemistry 40: 8563–8571,2001.

170. Kuczynski B, Reo NV. Evidence that plasmalogenis protective against oxidative stress in the ratbrain. Neurochem Res 31: 639–656, 2006.

171. Ladu MJ, Falduto MT, Manelli AM, Reardon CA,Getz GS, Frail DE. Isoform-specific binding ofapolipoprotein-E to beta-amyloid. J Biol Chem269: 23403–23406, 1994.

172. LaFerla FM. Calcium dyshomeostasis and intra-cellular signalling in Alzheimer’s disease. Nat RevNeurosci 3: 862–872, 2002.

173. Lal R, Lin H, Quist AP. Amyloid beta ion channel: 3Dstructure and relevance to amyloid channel para-digm. BBA Biomembranes 1768: 1966–1975, 2007.

174. Lashuel HA, Hartley D, Petre BM, Walz T, LansburyPT. Neurodegenerative disease: amyloid poresfrom pathogenic mutations. Nature 418: 291,2002.

175. Lashuel HA, Lansbury PT. Are amyloid diseasescaused by protein aggregates that mimic bacte-rial pore-forming toxins? Q Rev Biophys 39: 167–201, 2006.

176. Lau TL, Ambroggio EE, Tew DJ, Cappai R, Mas-ters CL, Fidelio GD, Barnham KJ, Separovic F.Amyloid-beta peptide disruption of lipid mem-branes and the effect of metal ions. J Mol Biol356: 759–770, 2006.

177. Lauderback CM, Hackett JM, Huang FF, KellerJN, Szweda LI, Markesbery WR, Butterfield DA.The glial glutamate transporter, GLT-1, is oxida-tively modified by 4-hydroxy-2-nonenal in theAlzheimer’s disease brain: the role of A beta1–42. J Neurochem 78: 413–416, 2001.

178. Lauderback CM, Kanski J, Hackett JM, Maeda N,Kindy MS, Butterfield DA. Apolipoprotein E mod-ulates Alzheimer’s A beta(1-42)-induced oxida-tive damage to synaptosomes in an allele-specificmanner. Brain Res 924: 90–97, 2002.

179. Lawson JA, Rokach J, FitzGerald GA. Isopros-tanes: formation, analysis and use as indices oflipid peroxidation in vivo. J Biol Chem 274:24441–24444, 1999.

180. Lee SH, Blair IA. Characterization of 4-oxo-2-non-enal as a novel product of lipid peroxidation.Chem Res Tox 13: 698–702, 2000.

181. Lee SJ, Liyanage U, Bickel PE, Xia WM, LansburyPT, Kosik KS. A detergent-insoluble membranecompartment contains A beta in vivo. Nat Med 4:730–734, 1998.

182. Lehtonen JYA, Holopainen JM, Kinnunen PKJ. Ac-tivation of phospholipase A(2) by amyloid beta-peptides in vitro. Biochemistry 35: 9407–9414,1996.

183. Leskovjan AC, Lanzirotti A, Miller LM. Amyloidplaques in PSAPP mice bind less metal thanplaques in human Alzheimer’s disease. Neuroim-age 47: 1215–1220, 2009.

184. Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG,Yang A, Gallagher M, Ashe KH. A specific amy-loid-beta protein assembly in the brain impairsmemory. Nature 440: 352–357, 2006.

185. Lim GP, Calon F, Morihara T, Yang FS, Teter B,Ubeda O, Salem N, Frautschy SA, Cole GM. Adiet enriched with the omega-3 fatty acid doco-sahexaenoic acid reduces amyloid burden in anaged Alzheimer mouse model. J Neurosci 25:3032–3040, 2005.

186. Lin H, Bhatia R, Lal R. Amyloid beta protein formsion channels: implications for Alzheimer’s diseasepathophysiology. FASEB J 15: 2433–2444, 2001.

187. Liu L, Komatsu H, Murray IVJ, Axelsen PH. Pro-motion of amyloid � protein misfolding and fibril-logenesis by a lipid oxidation product. J Mol Biol377: 1236–1250, 2008.

188. Liu SJ, Zukin RS. Ca2�-permeable AMPA recep-tors in synaptic plasticity and neuronal death.Trends Neurosci 30: 126–134, 2007.

189. Lorenzo A, Yuan ML, Zhang ZH, Paganetti PA,Sturchler-Pierrat C, Staufenbiel M, Mautino J, SolV, Sommer B, Yankner BA. Amyloid beta inter-acts with the amyloid precursor protein: a poten-tial toxic mechanism in Alzheimer’s disease. NatNeurosci 3: 460–464, 2000.

190. Lovell MA, Ehmann WD, Mattson MP, Markes-bery WR. Elevated 4-hydroxynonenal in ventric-ular fluid in Alzheimer’s disease. Neurobiol Aging18: 457–461, 1997.

191. Lovell MA, Robertson JD, Teesdale WJ, Camp-bell JL, Markesbery WR. Copper, iron and zinc inAlzheimer’s disease senile plaques. J Neurol Sci158: 47–52, 1998.

192. Lovell MA, Xie CS, Markesbery WR. Acrolein isincreased in Alzheimer’s disease brain and istoxic to primary hippocampal cultures. NeurobiolAging 22: 187–194, 2001.

193. Lukiw WJ, Cui JG, Marcheselli VL, Bodker M,Botkjaer A, Gotlinger K, Serhan CN, Bazan NG. Arole for docosahexaenoic acid-derived neuropro-tectin D1 in neural cell survival and Alzheimerdisease. J Clin Invest 115: 2774–2783, 2005.

194. Ma JY, Yee A, Brewer HB, Das S, Potter H. Am-yloid-associated proteins alpha(1)-antichymot-rypsin and apolipoprotein-E promote assemblyof Alzheimer beta-protein into filaments. Nature372: 92–94, 1994.

195. Maeba R, Sawada Y, Shimasaki H, Takahashi I,Ueta N. Ethanolamine plasmalogens protect cho-lesterol-rich liposomal membranes from oxida-tion caused by free radicals. Chem Phys Lipids120: 145–151, 2002.

196. Maeba R, Ueta N. Ethanolamine plasmalogensprevent the oxidation of cholesterol by reducingthe oxidizability of cholesterol in phospholipidbilayers. J Lipid Res 44: 164–171, 2003.

197. Mahley RW, Rall SC Jr. Apolipoprotein E: farmore than a lipid transport protein. Ann RevGenom Hum Gen 1: 507–537, 2000.

198. Manelli AM, Stine WB, Van Eldik LJ, Ladu MJ.ApoE A�1–42 interactions. J Mol Neurosci 23:235–246, 2004.

199. Marchesi VT. An alternative interpretation of theamyloid A beta hypothesis with regard to thepathogenesis of Alzheimer’s disease. Proc NatlAcad Sci USA 102: 9093–9098, 2005.

200. Marcus DL, Thomas C, Rodriguez C, SimberkoffK, Tsai JS, Strafaci JA, Freedman ML. Increasedperoxidation and reduced antioxidant enzymeactivity in Alzheimer’s disease. Exp Neurol 150:40–44, 1998.

201. Markesbery WR. Oxidative stress hypothesis inAlzheimer’s disease. Free Radic Biol Med 23:134–147, 1997.

202. Markesbery WR, Carney JM. Oxidative altera-tions in Alzheimer’s disease. Brain Pathol 9: 133–146, 1999.

203. Markesbery WR, Lovell MA. Four-hydroxynon-enal, a product of lipid peroxidation, is increasedin the brain in Alzheimer’s disease. NeurobiolAging 19: 33–36, 1998.

204. Martinez-Senac MD, Villalain J, Gomez-Fernan-dez JC. Structure of the Alzheimer beta-amyloidpeptide (25-35) and its interaction with nega-tively charged phospholipid vesicles. Eur JBiochem 265: 744–753, 1999.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 65

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 14: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

205. Matsuzaki K. Physicochemical interactions of am-yloid �-peptide with lipid bilayers. BBABiomembranes 1768: 1935–1942, 2007.

206. Matsuzaki K, Horikiri C. Interactions of amyloidbeta-peptide (1-40) with ganglioside-containingmembranes. Biochemistry 38: 4137–4142, 1999.

207. Matsuzaki K, Noguch T, Wakabayashi M, Ikeda K,Okada T, Ohashi Y, Hoshino M, Naiki H. Inhibi-tors of amyloid beta-protein aggregation medi-ated by GM1-containing raft-like membranes.BBA Biomembranes 1768: 122–130, 2007.

208. Mattson MP, Cheng B, Culwell AR, Esch FS,Lieberburg I, Rydel RE. Evidence for excitopro-tective and intraneuronal calcium-regulatingroles for secreted forms of the beta-amyloid pre-cursor protein. Neuron 10: 243–254, 1993.

209. Mattson MP, Cheng B, Davis D, Bryant K, Lieber-burg I, Rydel RE. �-Amyloid peptides destabilizecalcium homeostasis and render human corticalneurons vulnerable to excitotoxicity. J Neurosci12: 376–389, 1992.

210. McGrath LT, McGleenon BM, Brennan S, McCollD, McIlroy S, Passmore AP. Increased oxidativestress in Alzheimer’s disease as assessed with4-hydroxynonenal but not malondialdehyde.QJM 94: 485–490, 2001.

211. McIntosh LJ, Trush MA, Troncoso JC. Increasedsusceptibility of Alzheimer’s disease temporalcortex to oxygen free radical-mediated pro-cesses. Free Radic Biol Med 23: 183–190, 1997.

212. McLaurin J, Chakrabartty A. Membrane disrup-tion by Alzheimer beta-amyloid peptides medi-ated through specific finding to eitherphospholipids or gangliosides: implications forneurotoxicity. J Biol Chem 271: 26482–26489,1996.

213. McLaurin J, Chakrabartty A. Characterization ofthe interactions of Alzheimer beta-amyloid pep-tides with phospholipid membranes. Eur JBiochem 245: 355–363, 1997.

214. McLaurin J, Darabie AA, Morrison MR. Choles-terol, a modulator of membrane-associated Abeta-fibrillogenesis. Alz Dis Vasc Etiol Path 977:376–383, 2002.

215. McLaurin J, Franklin T, Fraser PE, ChakrabarttyA. Structural transitions associated with the in-teraction of Alzheimer beta-amyloid peptideswith gangliosides. J Biol Chem 273: 4506–4515,1998.

216. Minton AP. Implications of macromolecularcrowding for protein assembly. Curr Opin StructBiol 10: 34–39, 2000.

217. Miyata M, Smith JD. Apolipoprotein E allele-spe-cific antioxidant activity and effects on cytotox-icity by oxidative insults and beta-amyloidpeptides. Nat Gen 14: 55–61, 1996.

218. Molander-Melin M, Blennow K, Bogdanovic N,Dellheden B, Mansson JE, Fredman P. Structuralmembrane alterations in Alzheimer brains foundto be associated with regional disease develop-ment; increased density of gangliosides GM1and GM2 and loss of cholesterol in detergent-resistant membrane domains. J Neurochem 92:171–182, 2005.

219. Monji A, Utsumi H, Ueda T, Imoto T, Yoshida I,Hashioka S, Tashiro K, Tashiro N. The relation-ship between the aggregational state of the am-yloid-beta peptides and free radical generationby the peptides. J Neurochem 77: 1425–1432,2001.

220. Montine TJ, Neely MD, Quinn JF, Beal MF,Markesbery WR, Roberts LJ, Morrow JD. Lipidperoxidation in aging brain and Alzheimer’s dis-ease. Free Radic Biol Med 33: 620–626, 2002.

221. Montine TJ, Quinn JF, Milatovic D, Silbert LC,Dang T, Sanchez S, Terry E, Roberts LJ, Kaye JA,Morrow JD. Peripheral F-2-isoprostanes and F-4-neuroprostanes are not increased in Alzheimer’sdisease. Ann Neurol 52: 175–179, 2002.

222. Morandat S, Bortolato M, Anker G, Doutheau A,Lagarde M, Chauvet JP, Roux B. Plasmalogensprotect unsaturated lipids against UV-inducedoxidation in monolayer. Biochim Biophys Acta1616: 137–146, 2003.

223. Morris MC, Evans DA, Bienias JL, Tangney CC,Bennett DA, Wilson RS, Aggarwal N, Schneider J.Consumption of fish and n-3 fatty acids and riskof incident Alzheimer disease. Arch Neurol 60:940–946, 2003.

224. Morris MC, Evans DA, Tangney CC, Bienias JL,Wilson RS. Fish consumption and cognitive de-cline with age in a large community study. ArchNeurol 62: 1849–1853, 2005.

225. Morrow JD, Awad JA, Boss HJ, Blair IA, RobertsLJ, II. Non-cyclooxygenase-derived prostanoids(F2-Isoprostanes) are formed in situ on phosph-lipids. Proc Natl Acad Sci USA 89: 10721–10725,1992.

226. Morrow JD, Minton TA, Mukundan CR, CampbellMD, Zackert WE, Daniel VC, Badr KF, Blair IA,Roberts LJ II. Free radical-induced generation ofisoprostanes in vivo. J Biol Chem 269: 4317–4326, 1994.

227. Mulder C, Wahlund LO, Teerlink T, Blomberg M,Veerhuis R, van Kamp GJ, Scheltens P, SchefferPG. Decreased lysophosphatidylcholine/phos-phatidylcholine ratio in cerebrospinal fluid in Alz-heimer’s disease. J Neural Transm 110: 949–955,2003.

228. Munishkina LA, Cooper EM, Uversky VN, FinkAL. The effect of macromolecular crowding onprotein aggregation and amyloid fibril formation.J Mol Recognit 17: 456–464, 2004.

229. Murphy EJ, Huang HM, Cowburn RF, Lannfeltd L,Gibson GE. Phospholipid mass is increased infibroblasts bearing the Swedish amyloid precur-sor mutation. Brain Res Bull 69: 79–85, 2006.

230. Murphy RC. Free-radical-induced oxidation ofarachidonoyl plasmalogen phospholipids: antiox-idant mechanism and precursor pathway for bio-active eicosanoids. Chem Res Tox 14: 463–472,2001.

231. Murray IVJ, Liu L, Komatsu H, Uryu K, Xiao G,Lawson JA, Axelsen PH. Membrane mediatedamyloidogenesis and the promotion of oxidativelipid damage by amyloid beta proteins. J BiolChem 282: 9335–9345, 2007.

232. Murray IVJ, Sindoni ME, Axelsen PH. Promotionof oxidative lipid membrane damage by amyloidbeta proteins. Biochemistry 44: 12606–12613,2005.

233. Namba Y, Tomonaga M, Kawasaki H, Otomo E,Ikeda K. Apolipoprotein-E immunoreactivity incerebral amyloid deposits and neurofibrillary tan-gles in Alzheimers-disease and Kuru plaque am-yloid In Creutzfeldt-Jakob disease. Brain Res 541:163–166, 1991.

234. Natte R, Yamaguchi H, Maat-Schieman MLC,Prins FA, Neeskens P, Roos RAC, van Duinen SG.Ultrastructural evidence of early non-fibrillar Abeta 42 in the capillary basement membrane ofpatients with hereditary cerebral hemorrhagewith amyloidosis, Dutch type. Acta Neuropathol(Berl) 98: 577–582, 1999.

235. Nelson TJ, Alkon DL. Oxidation of cholesterol byamyloid precursor protein and beta-amyloid pep-tide. J Biol Chem 280: 7377–7387, 2005.

236. Nunomura A, Perry G, Pappolla MA, FriedlandRP, Hirai K, Chiba S, Smith MA. Neuronal oxida-tive stress precedes amyloid-beta deposition inDown syndrome. J Neuropath Exp Neurol 59:1011–1017, 2000.

237. Nurk E, Drevon CA, Refsum H, Solvoll K, VollsetSE, Nygard O, Nygaard HA, Engedal K, Tell GS,Smith AD. Cognitive performance among the el-derly and dietary fish intake: the HordalandHealth Study. Am J Clin Nutr 86: 1470–1478,2007.

238. O’Nuallain B, Shivaprasad S, Kheterpal I, WetzelR. Thermodynamics of A beta(1-40) amyloid fibrilelongation. Biochemistry 44: 12709–12718, 2005.

239. Okada T, Ikeda K, Wakabayashi M, Ogawa M,Matsuzaki K. Formation of toxic A beta(1-40) fi-brils on GM1 ganglioside-containing membranesmimicking lipid rafts: polymorphisms in A beta(1-40) fibrils. J Mol Biol 382: 1066–1074, 2008.

240. Okada T, Wakabayashi M, Ikeda K, MatsuzakiK. Formation of toxic fibrils of Alzheimer’s amy-loid beta-protein-(1-40) by monosialogangliosideGM1, a neuronal membrane component. J MolBiol 371: 481–489, 2007.

241. Opazo C, Huang XD, Cherny RA, Moir RD, Ro-her AE, White AR, Cappai R, Masters CL, TanziRE, Inestrosa NC, Bush AI. Metalloenzyme-likeactivity of Alzheimer’s disease beta-amyloid- Cu-de-pendent catalytic conversion of dopamine, choles-terol, and biological reducing agents to neurotoxicH2O2. J Biol Chem 277: 40302–40308, 2002.

242. Paltauf F. Ether lipids in biomembranes. ChemPhys Lipids 74: 101–139, 1994.

243. Pamplona R, Dalfo E, Ayala VR, Bellmunt MJ, PratJ, Ferrer I, Portero-Otin M. Proteins in humanbrain cortex are modified by oxidation, glycoxi-dation, and lipoxidation: effects of Alzheimer dis-ease and identification of lipoxidation targets. JBiol Chem 280: 21522–21530, 2005.

244. Patrono C, FitzGerald GA. Isoprostanes: poten-tial markers of oxidant stress in atherothrom-botic disease. Arterioscler Thromb Vasc Biol 17:2309–2315, 1997.

245. Permanne B, Perez C, Soto C, Frangione B, Wis-niewski T. Detection of apolipoprotein E dimericsoluble amyloid beta complexes in Alzheimer’sdisease brain supernatants. Biochem Biophys ResCommun 240: 715–720, 1997.

246. Petersen CAH, Alikhani N, Behbahani H, Wie-hager B, Pavlov PF, Alafuzoff I, Leinonen V, Ito A,Winblad B, Glaser E, Ankarcrona M. The amyloidbeta-peptide is imported into mitochondria viathe TOM import machinery and localized to mi-tochondrial cristae. Proc Natl Acad Sci USA 105:13145–13150, 2008.

247. Pettegrew JW, Panchalingam K, Hamilton RL,McClure RJ. Brain membrane phospholipid alter-ations in Alzheimer’s disease. Neurochem Res26: 771–782, 2001.

248. Pike CJ, RamezanArab N, Cotman CW. Beta-amyloid neurotoxicity in vitro: evidence of oxida-tive stress but not protection by antioxidants. JNeurochem 69: 1601–1611, 1997.

249. Pillot T, Goethals M, Vanloo B, Talussot C, Bras-seur R, Vandekerckhove J, Rosseneu M, Lins L.Fusogenic properties of the C-terminal domainof the Alzheimer beta-amyloid peptide. J BiolChem 271: 28757–28765, 1996.

250. Plourde M, Fortier M, Vandal M, Tremblay-Mercier J, Freemantle E, Begin M, Pifferi F,Cunnane SC. Unresolved issues in the link be-tween docosahexaenoic acid and Alzheimer’sdisease. Prost Leuk Essen Fatty Acids 77: 301–308, 2007.

251. Pollard HB, Arispe N, Rojas E. Ion channel hy-pothesis for Alzheimer amyloid peptide neuro-toxicity. Cell Molec Neurobiol 15: 513–526, 1995.

252. Prasad MR, Lovell MA, Yatin M, Dhillon H,Markesbery WR. Regional membrane phospho-lipid alterations in Alzheimer’s disease. Neuro-chem Res 23: 81–88, 1998.

253. Pratico D, Clark CM, Lee VMY, Trojanowski JQ,Rokach J, FitzGerald GA. Increased 8,12-iso-iPF(2 alpha)-VI in Alzheimer’s disease: correlationof a noninvasive index of lipid peroxidation withdisease severity. Ann Neurol 48: 809–812, 2000.

254. Pratico D, Delanty N. Oxidative injury in diseasesof the central nervous system: focus on Alzhei-mer’s disease. Am J Med 109: 577–585, 2000.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org66

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 15: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

255. Pratico D, Lee VMY, Trojanowski JQ, Rokach J,FitzGerald GA. Increased F-2-isoprostanes in Alz-heimer’s disease: evidence for enhanced lipidperoxidation in vivo. FASEB J 12: 1777–1783,1998.

256. Pratico D, Uryu K, Leight S, Trojanowswki JQ, LeeVMY. Increased lipid peroxidation precedes am-yloid plaque formation in an animal model ofAlzheimer amyloidosis. J Neurosci 21: 4183–4187, 2001.

257. Puglielli L, Friedlich AL, Setchell KDR, Nagano S,Opazo C, Cherny RA, Barnham KJ, Wade JD,Melov S, Kovacs DM, Bush AI. Alzheimer disease�-amyloid activity mimics cholesterol oxidase. JClin Invest 115: 2556–2563, 2005.

258. Qahwash IM, Boire A, Lanning J, Krausz T, PytelP, Meredith SC. Site-specific effects of peptidelipidation on beta-amyloid aggregation and cyto-toxicity. J Biol Chem 282: 36987–36997, 2007.

259. Quist A, Doudevski L, Lin H, Azimova R, Ng D,Frangione B, Kagan B, Ghiso J, Lal R. Amyloid ionchannels: a common structural link for protein-misfolding disease. Proc Natl Acad Sci USA 102:10427–10432, 2005.

260. Rajendran L, Schneider A, Schlechtingen G,Weidlich S, Ries J, Braxmeier T, Schwille P, SchulzJB, Schroeder C, Simons M, Jennings G, KnolkerHJ, Simons K. Efficient inhibition of the Alzhei-mer’s disease �-secretase by membrane target-ing. Science 320: 520–523, 2008.

261. Rangachari V, Moore BD, Reed DK, Sonoda LK,Bridges AW, Conboy E, Hartigan D, RosenberryTL. Amyloid-�(1-42) rapidly forms protofibrilsand oligomers by distinct pathways in low con-centrations of sodium dodecylsulfate. Biochem-istry 46: 12451–12462, 2007.

262. Reddy PH. Amyloid beta, mitochondrial struc-tural and functional dynamics in Alzheimer’s dis-ease. Exp Neurol 218: 286–292, 2009.

263. Refolo LM, Pappolla MA, Malester B, LaFrancoisJ, Bryant-Thomas T, Wang R, Tint GS, Sambam-urti K, Duff K. Hypercholesterolemia acceleratesthe Alzheimer’s amyloid pathology in a trans-genic mouse model. Neurobiol Dis 7: 321–331,2000.

264. Reiss D, Beyer K, Engelmann B. Delayed oxida-tive degradation of polyunsaturated diacyl phos-pholipids in the presence of plasmalogenphospholipids in vitro. Biochem J 323: 807–814,1997.

265. Relini A, Cavalleri O, Rolandi R, Gliozzi A. Thetwo-fold aspect of the interplay of amyloidogenicproteins with lipid membranes. Chem Phys Lipids158: 1–9, 2009.

266. Roberts LJ, Fessel JP. The biochemistry of theisoprostane, neuroprostane, and isofuran path-ways of lipid peroxidation. Chem Phys Lipids128: 173–186, 2004.

267. Roberts LJ, Montine TJ, Markesbery WR, TapperAR, Hardy P, Chemtob S, Dettbarn WD, MorrowJD. Formation of isoprostane-like compounds(neuroprostanes) in vivo from docosahexaenoicacid. J Biol Chem 273: 13605–13612, 1998.

268. Roberts LJ, II, Morrow JD. IsoProstanes: novelmarkers of endogenous lipid peroxidation andpotential mediators of oxidant injury. Ann NYAcad Sci 744: 237–242, 1994.

269. Roheim PS, Carey M, Forte T, Vega GL. Apolipo-proteins in human cerebrospinal fluid. Proc NatlAcad Sci USA 76: 4646–4649, 1979.

270. Rottkamp CA, Nunomura A, Raina AK, Sayre LM,Perry G, Smith MA. Oxidative stress, antioxi-dants, and Alzheimer disease. Alzheimer Dis As-soc Disord 14, Suppl 1: S62–S66, 2000.

271. Sanan DA, Weisgraber KH, Russell SJ, MahleyRW, Huang D, Saunders A, Schmechel D, Wis-niewski T, Frangione B, Roses AD, StrittmatterWJ. Apolipoprotein-E associates with beta-amy-loid peptide of Alzheimers-disease to form novelmonofibrils: isoform Apoe4 associates more Effi-ciently than Apoe3. J Clin Invest 94: 860–869,1994.

272. Sarell CJ, Syme CD, Rigby SEJ, Viles JH. Copper(II)binding to amyloid-beta fibrils of Alzheimer’s dis-ease reveals a picomolar affinity: stoichiometryand coordination geometry are independent of Abeta oligomeric form. Biochemistry 48: 4388–4402, 2009.

273. Sayre LM, Perry G, Harris PLR, Liu YH, SchubertKA, Smith MA. In situ oxidative catalysis by neu-rofibrillary tangles and senile plaques in Alzhei-mer’s disease: a central role for bound transitionmetals. J Neurochem 74: 270–279, 2000.

274. Sayre LM, Zelasko DA, Harris PLR, Perry G, Salo-mon RG, Smith MA. 4-Hydroxynonenal-derivedadvanced lipid peroxidation end products areincreased in Alzheimer’s disease. J Neurochem68: 2092–2097, 1997.

275. Schaefer EJ, Bongard V, Beiser AS, Lamon-FavaS, Robins SJ, Au R, Tucker KL, Kyle DJ, WilsonPWF, Wolf PA. Plasma phosphatidylcholine do-cosahexaenoic acid content and risk of dementiaand Alzheimer disease: the Framingham heartstudy. Arch Neurol 63: 1545–1550, 2006.

276. Schauerte JA, Wong PT, Wisser KC, Ding H, SteelDG, Gafni A. Simultaneous single molecule fluo-rescence and conductivity studies reveal distinctclasses of A� species on lipid bilayers. Biochem-istry 49: 3031–3039, 2010.

277. Schippling S, Kontush A, Arlt S, Buhmann C,Sturenburg HJ, Mann U, Muller-Thomsen T,Beisiegel U. Increased lipoprotein oxidation inAlzheimer’s disease. Free Radic Biol Med 28:351–360, 2000.

278. Schmechel DE, Saunders AM, Strittmatter WJ,Crain BJ, Huletter CM, Joo SH, Pericakvance MA,Goldgaber D, Roses AD. Increased amyloid beta-peptide deposition in cerebral-cortex as a conse-quence of apolipoprotein-E genotype in late-onset Alzheimer-disease. Proc Natl Acad Sci USA90: 9649–9653, 1993.

279. Schmitz C, Rutten BPF, Pielen A, Schafer S,Wirths O, Tremp G, Czech C, Blanchard V,Multhaup G, Rezaie P, Korr H, Steinbusch HWM,Pradier L, Bayer TA. Hippocampal neuron lossexceeds amyloid plaque load in a transgenicmouse model of Alzheimer’s disease. Am JPathol 164: 1495–1502, 2004.

280. Schneeweis LA, Koppaka V, Lund-Katz S, PhillipsMC, Axelsen PH. Structural analysis of lipopro-tein E particles. Biochemistry 44: 12525–12534,2005.

281. Sengupta P, Garai K, Sahoo B, Shi Y, CallawayDJE, Maiti S. The amyloid beta peptide [A beta(1-40)] is thermodynamically soluble at physiologicalconcentrations. Biochemistry 42: 10506–10513,2003.

282. Shankar GM, Li SM, Mehta TH, Garcia-Munoz A,Shepardson NE, Smith I, Brett FM, Farrell MA,Rowan MJ, Lemere CA, Regan CM, Walsh DM,Sabatini BL, Selkoe DJ. Amyloid-beta proteindimers isolated directly from Alzheimer’s brainsimpair synaptic plasticity and memory. Nat Med14: 837–842, 2008.

283. Shao H, Jao S, Ma K, Zagorski MG. Solutionstructures of micelle-bound amyloid beta-(1-40)and beta-(1-42) peptides of Alzheimer’s disease.J Mol Biol 285: 755–773, 1999.

284. Shearer J, Szalai VA. The Amyloid-� peptide ofAlzheimer’s disease binds Cu� in a linear Bis-Hiscoordination environment: insight into a possibleneuroprotective mechanism for the amyloid-�peptide. J Am Chem Soc 130: 17826–17835,2008.

285. Shin SJ, Lee SE, Boo JH, Kim M, Yoon YD, Kim SI,Mook-Jung I. Profiling proteins related to amy-loid deposited brain of Tg2576 mice. Proteomics4: 3359–3368, 2004.

286. Shringarpure R, Grune T, Sitte N, Davies KJA.4-Hydroxynonenal-modified amyloid-beta pep-tide inhibits the proteasome: possible impor-tance in Alzheimer’s disease. Cell Mol Life Sci 57:1802–1809, 2000.

287. Siegel SJ, Bieschke J, Powers ET, Kelly JW. Theoxidative stress metabolite 4-hydroxynonenalpromotes Alzheimer protofibril formation. Bio-chemistry 46: 1503–1510, 2007.

288. Simons M, Keller P, De Strooper B, BeyreutherK, Dotti CG, Simons K. Cholesterol depletioninhibits the generation of beta-amyloid in hip-pocampal neurons. Proc Natl Acad Sci USA 95:6460–6464, 1998.

289. Sjogren M, Blennow K. The link between choles-terol and Alzheimer’s disease. World J Biol Psych6: 85–97, 2005.

290. Smith DP, Ciccotosto GD, Tew DJ, Fodero-Tavo-letti MT, Johanssen T, Masters CL, Barnham KJ,Cappai R. Concentration dependent Cu2� in-duced aggregation and dityrosine formation ofthe Alzheimer’s disease amyloid-� peptide. Bio-chemistry 46: 2881–2891, 2007.

291. Smith DP, Smith DG, Curtain CC, Boas JF, Pil-brow JR, Ciccotosto GD, Lau TL, Tew DJ, PerezK, Wade JD, Bush AI, Drew SC, Separovic F,Masters CL, Cappai R, Barnham KJ. Copper me-diated amyloid-beta toxicity is associated with anintermolecular histidine bridge. J Biol Chem 281:15145–15154, 2006.

292. Smith MA, Casadesus G, Joseph JA, Perry G.Amyloid-beta and tau serve antioxidant functionsin the aging and Alzheimer brain. Free Radic BiolMed 33: 1194–1199, 2002.

293. Smith MA, Harris PLR, Sayre LM, Beckman JS,Perry G. Widespread peroxynitrite-mediateddamage in Alzheimer’s disease. J Neurosci 17:2653–2657, 1997.

294. Smith MA, Hirai K, Hsiao K, Pappolla MA, HarrisPLR, Siedlak SL, Tabaton M, Perry G. Amyloid-beta deposition in Alzheimer transgenic mice isassociated with oxidative stress. J Neurochem70: 2212–2215, 1998.

295. Smith MA, Perry G, Richey PL, Sayre LM, Ander-son VE, Beal MF, Kowall N. Oxidative damage inAlzheimer’s. Nature 382: 120–121, 1996.

296. Smith MA, Taneda S, Richey PL, Miyata S, YanSD, Stern D, Sayre LM, Monnier VM, Perry G.Advanced Maillard reaction end-products are as-sociated with Alzheimer-disease pathology. ProcNatl Acad Sci USA 91: 5710–5714, 1994.

297. Sokolov Y, Kozak JA, Kayed R, Chanturiya A,Glabe C, Hall JE. Soluble amyloid oligomers in-crease bilayer conductance by altering dielectricstructure. J Gen Physiol 128: 637–647, 2006.

298. Song WL, Lawson JA, Reilly D, Rokach J, ChangCT, Giasson B, FitzGerald GA. Neurofurans,novel indices of oxidant stress derived from do-cosahexaenoic acid. J Biol Chem 283: 6–16,2008.

299. Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM,Ingelsson M, Hyman B, Burton MA, Goldstein LE,Duong S, Tanzi RE, Moir RD. The Alzheimer’sdisease-associated amyloid �-protein is an anti-microbial peptide. PLoS ONE 5: e9505, 2010.

300. Sparks DL, Scheff SW, Hunsaker JC, Liu HC,Landers T, Gross DR. Induction of Alzheimer-likebeta-amyloid immunoreactivity in the brains ofrabbits with dietary-cholesterol. Exp Neurol 126:88–94, 1994.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 67

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 16: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

301. Stewart CR, Wilson LM, Zhang Q, Pham CLL,Waddington LJ, Staples MK, Stapleton D, KellyJW, Howlett GJ. Oxidized cholesterol metabo-lites found in human atherosclerotic lesions pro-mote apolipoprotein C-II amyloid fibril formation.Biochemistry 46: 5552–5561, 2007.

302. Stokin GB, Lillo C, Falzone TL, Brusch RG, Rock-enstein E, Mount SL, Raman R, Davies P, MasliahE, Williams DS, Goldstein LSB. Axonopathy andtransport deficits early in the pathogenesis ofAlzheimer’s disease. Science 307: 1282–1288,2005.

303. Stratman NC, Castle CK, Taylor BM, Epps DE,Melchior GW, Carter DB. Isoform-specific inter-actions of human apolipoprotein E to an interme-diate conformation of human Alzheimer amyloid-beta peptide. Chem Phys Lipids 137: 52–61,2005.

304. Strausak D, Mercer JFB, Dieter HH, Stremmel W,Multhaup G. Copper in disorders with neurolog-ical symptoms: Alzheimer’s, Menkes, and Wilsondiseases. Brain Res Bull 55: 175–185, 2001.

305. Streltsov VA. X-ray absorption and diffractionstudies of the metal binding sites in amyloid be-ta-peptide. Eur Biophys J 37: 257–263, 2008.

306. Streltsov VA, Titmuss SJ, Epa VC, Barnham KJ,Masters CL, Varghese JN. The structure of theamyloid-beta peptide high-affinity copper IIbinding site in Alzheimer disease. Biophys J 95:3447–3456, 2008.

307. Strittmatter WJ, Weisgraber KH, Huang DY,Dong LM, Salvesen GS, Pericakvance M, Sch-mechel D, Saunders AM, Goldgaber D, RosesAD. Binding of human apolipoprotein-E to syn-thetic amyloid-beta peptide: isoform-specific ef-fects and implications for late-onset Alzheimer-disease. Proc Natl Acad Sci USA 90: 8098–8102,1993.

308. Subasinghe S, Unabia S, Barrow CJ, Mok SS,Aguilar MI, Small DH. Cholesterol is necessaryboth for the toxic effect of A beta peptides onvascular smooth muscle cells and for A beta bind-ing to vascular smooth muscle cell membranes. JNeurochem 84: 471–479, 2003.

309. Sultana R, Butterfield DA. Oxidatively modifiedGST and MRP1 in Alzheimer’s disease brain: im-plications for accumulation of reactive lipid per-oxidation products. Neurochem Res 29: 2215–2220, 2004.

310. Surewicz WK, Mantsch HH. The conformation ofdynorphin A-(1-13) in aqueous solution as stud-ied by Fourier transform infrared spectroscopy. JMol Struct 214, 143–147, 1989.

311. Svennerholm L, Gottfries CG. Membrane-lipids,selectively diminished in Alzheimer brains, suggestsynapse loss as a primary event in early-onset form(type-I) and demyelination in late-onset form (type-II). J Neurochem 62: 1039–1047, 1994.

312. Tamaoka A, Miyatake F, Matsuno S, Ishii K, Na-gase S, Sahara N, Ono S, Mori H, Wakabayashi K,Tsuji S, Takahashi H, Shoji S. Apolipoprotein Eallele-dependent antioxidant activity in brainswith Alzheimer’s disease. Neurol 54: 2319–2321,2000.

313. Terzi E, Holzemann G, Seelig J. Alzheimer beta-amyloid peptide 25-35: electrostatic interactionswith phospholipid membranes. Biochemistry 33:7434–7441, 1994.

314. Terzi E, Holzemann G, Seelig J. Self-associationof beta-amyloid peptide (1-40) in solution andbinding to lipid membranes. J Mol Biol 252: 633–642, 1995.

315. Terzi E, Holzemann G, Seelig J. Interaction of Alz-heimer beta-amyloid peptide(1-40) with lipidmembranes. Biochemistry 36: 14845–14852,1997.

316. Tickler AK, Smith DG, Ciccotosto GD, Tew DJ,Curtain CC, Carrington D, Masters CL, Bush AI,Cherny RA, Cappai R, Wade JD, Barnham KJ.Methylation of the imidazole side chains of theAlzheimer disease amyloid-� peptide results inabolition of superoxide dismutase-like structuresand inhibition of neurotoxicity. J Biol Chem 280:13355–13363, 2005.

317. Tien M, Berlett BS, Levine RL, Chock PB, Stadt-man ER. Peroxynitrite-mediated modification ofproteins at physiological carbon dioxide concen-tration: pH dependence of carbonyl formation,tyrosine nitration, and methionine oxidation.Proc Natl Acad Sci USA 96: 7809–7814, 1999.

318. Tomiyama T, Matsuyama S, Iso H, Umeda T, Ta-kuma H, Ohnishi K, Ishibashi K, Teraoka R,Sakama N, Yamashita T, Nishitsuji K, Ito K, Shi-mada H, Lambert MP, Klein WL, Mori H. A mousemodel of amyloid � oligomers: their contributionto synaptic alteration, abnormal tau phosphory-lation, glial activation, and neuronal loss in vivo. JNeurosci 30: 4845–4856, 2010.

319. Torp R, Head E, Milgram NW, Hahn F, OttersenOP, Cotman CW. Ultrastructural evidence offibrillar beta-amyloid associated with neuronalmembranes in behaviorally characterized ageddog brains. Neuroscience 96: 495–506, 2000.

320. Townsend M, Shankar GM, Mehta T, Walsh DM,Selkoe DJ. Effects of secreted oligomers of am-yloid beta-protein on hippocampal synaptic plas-ticity: a potent role for trimers. J Physiol 572:477–492, 2006.

321. Treiber C, Quadir MA, Voigt P, Radowski M, XuSJ, Munter LM, Bayer TA, Schaefer M, Haag R,Multhaup G. Cellular copper import by nanocar-rier systems, intracellular availability, and effectson amyloid beta peptide secretion. Biochemistry48: 4273–4284, 2009.

322. Trojanowski JQ, Lee VM. The role of tau in Alz-heimer’s disease. Med Clin North Am 86: 615–627, 2002.

323. Tully AM, Roche HM, Doyle R, Fallon C, Bruce I,Lawlor B, Coakley D, Gibney MJ. Low serumcholesteryl ester-docosahexaenoic acid levels inAlzheimer’s disease: a case-control study. Br JNutr 89: 483–489, 2003.

324. Uchida K. Current status of acrolein as a lipidperoxidation product. Trends Cardiovasc Med 9:109–113, 1999.

325. Uchida K. 4-Hydroxy-2-nonenal: a product andmediator of oxidative stress. Prog Lipid Res 42:318–343, 2003.

326. Uchida K, Kanematsu M, Morimitsu Y, Osawa T,Noguchi N, Niki E. Acrolein is a product of lipidperoxidation reaction: formation of free acroleinand its conjugate with lysine residues in oxidizedlow density lipoproteins. J Biol Chem 273:16058–16066, 1998.

327. Uchida K, Kanematsu M, Sakai K, Matsuda T,Hattori N, Mizuno Y, Suzuki D, Miyata T, NoguchiN, Niki E, Osawa T. Protein-bound acrolein: po-tential markers for oxidative stress. Proc NatlAcad Sci USA 95: 4882–4887, 1998.

328. Uchida K, Stadtman ER. Modification of histidine-residues in proteins by reaction with 4-hy-droxynonenal. Proc Natl Acad Sci USA 89: 4544–4548, 1992.

329. Usui K, Hulleman JD, Paulsson JF, Siegel SJ, Pow-ers ET, Kelly JW. Site-specific modification ofAlzheimer’s peptides by cholesterol oxidationproducts enhances aggregation energetics andneurotoxicity. Proc Natl Acad Sci USA 106:18563–18568, 2009.

330. Van Nostrand WE, Melchor JP, Ruffini L. Patho-logic amyloid beta-protein cell surface fibril as-sembly on cultured human cerebrovascularsmooth muscle cells. J Neurochem 70: 216–223,1998.

331. Varadarajan S, Yatin S, Aksenova M, ButterfieldDA. Alzheimer’s amyloid beta-peptide-associ-ated free radical oxidative stress and neurotox-icity. J Struct Biol 130: 184–208, 2000.

332. Varadarajan S, Yatin S, Butterfield DA. Alzhei-mer’s amyloid beta peptide (1-42) fibrils are notalways neurotoxic. Alzheimers Rep 3: 71–76,2000.

333. Wahrle SE, Jiang H, Parsadanian M, Hartman RE,Bales KR, Paul SM, Holtzman DM. Deletion ofAbca1 increases A beta deposition in the PDAPPtransgenic mouse model of Alzheimer disease. JBiol Chem 280: 43236–43242, 2005.

334. Wakabayashi M, Matsuzaki K. Formation of amy-loids by a beta-(1-42) on NGF-differentiatedPC12 cells: roles of gangliosides and cholesterol.J Mol Biol 371: 924–933, 2007.

335. Wakabayashi M, Okada T, Kozutsumi Y, Matsu-zaki K. GM1 ganglioside-mediated accumulationof amyloid beta-protein on cell membranes.Biochem Biophys Res Commun 328: 1019–1023,2005.

336. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK,Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Nat-urally secreted oligomers of amyloid beta proteinpotently inhibit hippocampal long-term potenti-ation in vivo. Nature 416: 535–539, 2002.

337. Walsh DM, Lomakin A, Benedek GB, CondronMM, Teplow DB. Amyloid beta-protein fibrillo-genesis: detection of a protofibrillar intermedi-ate. J Biol Chem 272: 22364–22372, 1997.

338. Wang SSS, Rymer DL, Good TA. Reduction incholesterol and sialic acid content protects cellsfrom the toxic effects of beta-amyloid peptides.J Biol Chem 276: 42027–42034, 2001.

339. Waschuk SA, Elton EA, Darabie AA, Fraser PE,McLaurin J. Cellular membrane composition de-fines Abeta-lipid interactions. J Biol Chem 276:33561–33568, 2001.

340. Weisgraber KH, Shinto LH. Identification of thedisulfide-linked homodimer of apolipoprotein E3in plasma: impact on receptor-binding activity. JBiol Chem 266: 12029–12034, 1991.

341. Widenbrant MJO, Rajadas J, Sutardja C, FullerGG. Lipid-induced �-amyloid peptide assem-blage fragmentation. Biophys J 91: 4071–4080,2006.

342. Williams TI, Lynn BC, Markesbery WR, Lovell MA.Increased levels of 4-hydroxynonenal and acro-lein, neurotoxic markers of lipid peroxidation, inthe brain in mild cognitive impairment and earlyAlzheimer’s disease. Neurobiol Aging 27: 1094–1099, 2006.

343. Wisniewski T, Castano EM, Golabek A, Vogel T,Frangione B. Acceleration of Alzheimers fibril for-mation by apolipoprotein-E in-vitro. Am J Pathol145: 1030–1035, 1994.

344. Wong PT, Schauerte JA, Wisser KC, Ding H, LeeEL, Steel DG, Gafni A. Amyloid-beta membranebinding and permeabilization are distinct pro-cesses influenced separately by membranecharge and fluidity. J Mol Biol 386: 81–96, 2009.

345. Wu Y, Luo Y. Transgenic C. elegans as a model inAlzheimer’s research. Curr Alzheimer Res 2: 37–47, 2005.

346. Yamaguchi H, Maat-Schieman MLC, van DuinenSG, Prins FA, Neeskens P, Natte R, Roos RAC.Amyloid beta protein (A beta) starts to deposit asplasma membrane-bound form in diffuse plaquesof brains from hereditary cerebral hemorrhagewith amyloidosis-Dutch type, Alzheimer diseaseand nondemented aged subjects. J NeuropathExp Neurol 59: 723–732, 2000.

347. Yamazaki T, Chang TY, Haass C, Ihara Y. Accu-mulation and aggregation of amyloid beta-pro-tein in late endosomes of Niemann-Pick type Ccells. J Biol Chem 276: 4454–4460, 2001.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org68

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from

Page 17: Oxidative Stress and Cell Membranes in the Pa Tho Genesis of DA

348. Yanagisawa K, Odaka A, Suzuki N, Ihara Y. GM1ganglioside-bound amyloid beta-protein (A be-ta): a possible form of preamyloid in Alzheimer’sdisease. Nat Med 1: 1062–1066, 1995.

349. Yang DS, Smith JD, Zhou ZM, Gandy SE, MartinsRN. Characterization of the binding of amyloid-beta peptide to cell culture-derived native apoli-poprotein E2, E3, and E4 isoforms and toisoforms from human plasma. J Neurochem 68:721–725, 1997.

350. Yao JK, Wengenack TM, Curran GL, Poduslo JF.Reduced membrane lipids in the cortex of Alz-heimer’s disease transgenic mice. NeurochemRes 34: 102–108, 2009.

351. Yao ZX, Drieu K, Szweda LI, Papadopoulos V.Free radicals and lipid peroxidation do not me-diate beta-amyloid-induced neuronal cell death.Brain Res 847: 203–210, 1999.

352. Yip CM, Darabie AA, McLaurin J. A beta 42-peptide assembly on lipid bilayers. J Mol Biol318: 97–107, 2002.

353. Yip CM, McLaurin J. Amyloid-beta peptide as-sembly: a critical step in fibrillogenesis and mem-brane disruption. Biophys J 80: 1359–1371,2001.

354. Yoshiike Y, Kayed R, Milton SC, Takashima A,Glabe CG. Pore-forming proteins share structuraland functional homology with amyloid oligomers.Neuromol Med 9: 270–275, 2007.

355. Yoshiike Y, Tanemura K, Murayama O, Akagi T,Murayama M, Sato S, Sun XY, Tanaka N,Takashima A. New insights on how metals dis-rupt amyloid beta-aggregation and their effectson amyloid-beta cytotoxicity. J Biol Chem 276:32293–32299, 2001.

356. Yoshimoto N, Tasaki M, Shimanouchi T, Umako-shi H, Kuboi R. Oxidation of cholesterol catalyzedby amyloid beta-peptide (A beta)-Cu complex onlipid membrane. J Biosci Bioeng 100: 455–459,2005.

357. Zagol-Ikapitte I, Masterson TS, Amarnath V,Montine TJ, Andreasson KI, Boutaud O, OatesJA. Prostaglandin H-2-derived adducts of pro-teins correlate with Alzheimer’s disease severity.J Neurochem 94: 1140–1145, 2005.

358. Zaman Z, Roche S, Fielden P, Frost PG, NiriellaDC, Cayley AC. Plasma concentrations of vita-mins A and E and carotenoids in Alzheimer’sdisease. Age Ageing 21: 91–94, 1992.

359. Zerbinatti CV, Wahrle SE, Kim H, Cam JA, BalesK, Paul SM, Holtzman DM, Bu GJ. ApolipoproteinE and low density lipoprotein receptor-relatedprotein facilitate intraneuronal A beta 42 accu-mulation in amyloid model mice. J Biol Chem281: 36180–36186, 2006.

360. Zhang QH, Powers ET, Nieva J, Huff ME, DendleMA, Bieschke J, Glabe CG, Eschenmoser A,Wentworth P, Lerner RA, Kelly JW. Metabolite-initiated protein misfolding may trigger Alzhei-mer’s disease. Proc Natl Acad Sci USA 101:4752–4757, 2004.

361. Zhou S, Zhou H, Walian PJ, Jap BK. Regulation of�-secretase activity in Alzheimer’s disease. Bio-chemistry 46: 2553–2563, 2007.

362. Zommara M, Tachibana N, Mitsui K, Nakatani N,Sakono M, Ikeda I, Imaizumi K. Inhibitory effectof ethanolamine plasmalogen on iron-dependentand copper-dependent lipid-peroxidation. FreeRadic Biol Med 18: 599–602, 1995.

363. Zou K, Gong JS, Yanagisawa K, Michikawa M. Anovel function of monomeric amyloid beta-pro-tein serving as an antioxidant molecule againstmetal-induced oxidative damage. J Neurosci 22:4833–4841, 2002.

364. Zubenko GS. Hippocampal membrane-alterationin Alzheimers-disease. Brain Res 385: 115–121,1986.

REVIEWS

PHYSIOLOGY • Volume 26 • February 2011 • www.physiologyonline.org 69

on January 24, 2012physiologyonline.physiology.org

Dow

nloaded from