Вы находитесь на странице: 1из 17

Oxidative Stress and Cell Membranes in the Pathogenesis of Alzheimer's Disease

Paul H. Axelsen, Hiroaki Komatsu and Ian V. J. Murray


Physiology 26:54-69, 2011. doi:10.1152/physiol.00024.2010 You might find this additional info useful... This article cites 363 articles, 102 of which can be accessed free at: http://physiologyonline.physiology.org/content/26/1/54.full.html#ref-list-1 This article has been cited by 1 other HighWire hosted articles Intra-membrane Oligomerization and Extra-membrane Oligomerization of Amyloid- Peptide Are Competing Processes as a Result of Distinct Patterns of Motif Interplay Yi-Jiong Zhang, Jing-Ming Shi, Cai-Juan Bai, Han Wang, Hai-Yun Li, Yi Wu and Shang-Rong Ji J. Biol. Chem., January 2, 2012; 287 (1): 748-756. [Abstract] [Full Text] [PDF]
Downloaded from physiologyonline.physiology.org on January 24, 2012

Updated information and services including high resolution figures, can be found at: http://physiologyonline.physiology.org/content/26/1/54.full.html Additional material and information about Physiology can be found at: http://www.the-aps.org/publications/physiol

This infomation is current as of January 24, 2012.

Physiology (formerly published as News in Physiological Science) publishes brief review articles on major physiological developments. It is published bimonthly in February, April, June, August, October, and December by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright 2011 by the American Physiological Society. ISSN: 1548-9213, ESSN: 1548-9221. Visit our website at http://www.the-aps.org/.

REVIEWS

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

Oxidative Stress and Cell Membranes in the Pathogenesis of Alzheimers Disease


Amyloid proteins and oxidative stress are believed to have central roles in the development of Alzheimers 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 Alzheimers disease pathogenesis is discussed in which lipid membranes assume both roles and promote the conversion of monomeric amyloid proteins into

Paul H. Axelsen,1,2 Hiroaki Komatsu,1 and 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 Science Center, College Station, Texas

Downloaded from physiologyonline.physiology.org on January 24, 2012

brils, the pathognomonic histopathological lesion of the disease.


The literature of Alzheimers disease (AD) research is vast, complex, and often contradictory. Nevertheless, a broad perspective on this literature, and on what typically does and does not happen in biological systems, suggests either that lipid membrane damage is directly involved in the pathogenesis of AD or that it is an important consequence of AD. Therefore, investigations into the relationship between membrane damage and amyloidogenesis may yield important insights into AD pathogenesis. The amyloid hypothesis has driven much of the research on AD pathogenesis since it was proposed in 1991 (123, 124). In its simplest form, this hypothesis suggests that the accumulation of amyloid beta (A ) proteins in brain tissue drives the pathogenesis of AD. A proteins originate from a large transmembrane protein of unknown function known as amyloid precursor protein (APP) by the action of -secretase and -secretase activity (FIGURE 1). The specic cleavage site of -secretase is somewhat variable, yielding proteins ranging from 39 to 43 residues in length, with 40 and 42 residue forms predominating (A 40 and A 42). The non-amyloidogenic pathway for APP processing involves -secretases, a family of enzymes that cleave APP near the middle of the A protein segment. The amyloid hypothesis was initially based on genetic studies of familial AD and the occurrence of AD-like pathology in Down Syndrome. Among the most cited experimental studies in support of this hypothesis are those in which A proteins impair physiological and cognitive function when injected into rodent brains (72, 136, 184, 282, 320, 336). Nevertheless, the amyloid hypothesis has been criticized for being incomplete and vague. For example, it is unable to explain nonfamilial or sporadic cases of AD (the most prevalent form), how bril formation leads to neuronal death, the poor clinicopathological correlation between amyloid plaque burden and cognitive impairment (279), the hyperphosphorylation and aggregation of tau proteins as neurobrillary tangles, and axonopathies that precede amyloid deposition (302). These criticisms notwithstanding, there are no alternative hypotheses with nearly as much support, and the amyloid hypothesis is readily adapted to answer some of these criticisms. For example, the pathologies that precede amyloid deposition may be due to prebrillar intermediate forms of A protein, and plaques may represent an inert form of the protein (62, 125, 337). Supporting this idea is the observation that a variant form of A that oligomerizes but does not brillize can nonetheless cause several pathological manifestations of AD other than bril formation (318). It has been suggested that there is a tenuous balance among the rates of A protein production, aggregation, and elimination in brain tissue, such that a small disturbance applied over sufcient time causes the pathological accumulation of A protein as amyloid brils in AD. However, the concentrations of A proteins in the brain appear to be far lower than their aqueous in vitro solubility (143, 238, 281). Therefore, A proteins should not form amyloid brils in the brain even if concentrations are modestly elevated. Yet, brils do form with advancing age in virtually everyone, indicating that factors other than A protein concentration must have a role in causing them to form. If small changes in protein concentration cannot account for bril formation, then we must consider factors

54

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

REVIEWS
such as macromolecular crowding (216, 228), locally high concentrations of A proteins at synapses (71), chemical modication (30, 161, 231, 287, 360), cross-linking (17, 22, 290, 291), or metal complex formation (6, 31, 92, 105, 153, 272, 284, 306). Understanding the chemical mechanisms that reduce the solubility of A proteins in brain tissue is key to understanding why AD pathology develops in some individuals and not others. as an intact methionine side chain is present (5, 21, 45, 47, 49, 232). A proteins and amyloid plaques bind redoxactive transition metals that are likely to be the actual site of redox activity (76, 273). Copper levels are signicantly increased in amyloid plaques (183, 191, 304), although comparatively subtle increases in copper transport across cell membranes may cause signicant changes in A protein turnover (321). Excess dietary copper increases AD-like pathology in a mouse model (160). Cu2 and Zn2 chelators appear to inhibit A deposition in the brains of mouse models (64), and metal depletion promotes the disaggregation of A brils (65). Cu2 potentiates the neurotoxicity of A 42 A 40 in embryonic rodent neurons, and its effect is mediated by H2O2 (135, 355). Studies of copper in the pathogenesis of AD have been extensively reviewed (39, 90, 100, 305). In contrast, Zn2 ions are redoxinert and able to protect/rescue human cells in tissue culture from A and Cu2 toxicity (75). A recent study, however, observed that the zinc released at synapses with neurotransmitters caused the accumulation of toxic A oligomers to these membranes (86). It has been suggested that A proteins split into fragments that are both neurotoxic and able to generate additional oxygen radicals (48, 128), although these ndings have been strongly refuted (89). Nevertheless, electron paramagnetic resonance spectroscopy has shown that there is a strong correlation between the intensity of radical generation by A and neurotoxicity (219). In these studies, preincubation of A to form brils increased its toxicity. In contrast, replacing the redox-active sulfur atom in residue Met35 with methylene (CH2) resulted in a peptide that formed brillar structures but had no demonstrable toxicity toward cultured hippocampal neurons. In the same experimental system, vitamin E (presumably acting as an antioxidant) neutralized the neurotoxicity of A but had no effect on its ability to form

Oxidative Stress
Oxidative stress is caused by a dense, complex and heterogeneous network of oxidizing reactions running counter to the reducing conditions that otherwise prevail in cells and tissues. It has been suggested that the accumulation of A proteins in the brain may be a protective response to oxidative stress (16, 28, 162, 164, 273, 292). The density of plaques containing A protein correlates inversely with markers of oxidative damage (75), and the cortical deposition of A proteins correlates with reduced oxidative damage in Down syndrome (236). Moreover, A proteins prevent lipoprotein oxidation (163) and metal-induced neuronal death in culture (363). However, it is inherently difcult to quantify oxidative stress, and attempts to do this are subject to many methodological pitfalls. For example, a study concluding that the primary mechanism of A toxicity does not involve oxidative pathways measured thiobarbituric acid reactive substances (TBARS) to assess lipid peroxidation (248). However, assays for TBARS measure only a small subset of the diverse oxidation products generated, as well as substances not derived from lipids. Other studies suggesting that A has antioxidant activity were not controlled for the inherent redox activity of peptide groups or conducted in the presence of biologically relevant electron donors and acceptors to drive the reactions (23, 24). Overall, it seems more likely that A proteins promote oxidative stress (42, 50, 91, 201, 202, 254). The brain in AD appears to sustain more oxidative damage than normal brain (200, 211, 331), exhibits an increased susceptibility to oxidative stress (190, 220, 277), and has relatively low levels of naturally occurring antioxidants such as -tocopherol (144, 358). Regions of the brain rich in A proteins also have increased levels of protein oxidation (129). The overexpression of A proteins in transgenic mice, in C. elegans, and in cell culture, increases biomarkers of oxidative stress (256, 345). A proteins cause H2O2 and lipid peroxides to accumulate in cells (27). Catalase protects cells from A toxicity, and cell lines selected for resistance to A toxicity also become resistant to the cytotoxic action of H2O2. A proteins promote the oxidation of compounds such as dopamine, phospholipids, and cholesterol (40, 134, 135, 235, 241, 257, 356) as long

Downloaded from physiologyonline.physiology.org on January 24, 2012

FIGURE 1. Processing of the amyloid precursor protein


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

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

55

REVIEWS
brils (332). Another study concluding that free radicals do not mediate A -induced neurotoxicity used ginkgolides (a purported anti-oxidant component of Ginkgo biloba leaves) and vitamin E to inhibit oxidation (351). Although neither agent protected cells from apoptosis and death, it is unlikely that they completely halted radical-mediated oxidative damage. Proteomics studies of oxidative stress have focused 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 taken for evidence of damage by oxidation, and the nature of the protein modication is not explicitly dened. Two studies have examined the ability of A proteins to induce oxidative modications in other proteins (34), and A proteins may also undergo oxidative damage, particularly His, Tyr, and Met side chains (15, 32, 43, 132, 139). However, it is difcult to imagine how oxidative damage to proteins, especially A proteins, would lower the aqueous solubility of A proteins and promote bril formation. type of link is overwhelming. A proteins are produced by cleavage of APP at two sites, one site being located approximately at the midpoint of the transmembrane segment (FIGURE 1). As a consequence, the COOH-terminal residues of A proteins that were part of the APP transmembrane segment are uniformly hydrophobic. Following their cleavage from APP, some investigators have observed that A proteins remain associated with detergent-resistant lipid membrane domains in the brain (181), or with membrane-anchored APP (189). It has also been hypothesized that A proteins bind to the transmembrane helices of membrane proteins and cause their dysfunction (199). Ultrastructural studies suggest that amyloid bril formation tends to occur rst in portions of diffuse amyloid deposits that are closest to membranes (234, 319, 346). A 40 with the E22Q mutation (responsible for hereditary cerebral hemorrhage with amyloidosis-Dutch type) will brillize on the surface membrane of human cerebrovascular smooth muscle cells (330). Despite a substantial hydrophobic segment, the general conclusion reached by most investigators is that A proteins have little afnity for neutral lipid membranes (205). Techniques such as the hydration of a mixed protein-lipid lm must be used to induce the penetration of A proteins into a neutral lipid bilayer (83). One laboratory investigating A 40 and another investigating A 42 have documented that the proteins situate differently in membranes depending on whether they are embedded in a bilayer membrane or allowed to associate with the surface of a preformed membrane (33, 106, 176). In general, anionic lipids tend to induce A proteins to adopt extended structure (35, 63, 66, 76, 130, 167169, 204, 213, 214, 313, 314, 344, 352). Possible reasons for the inducement of structure and bril formation by protein-lipid interaction have been reviewed, including the ability of such interactions to serve as templates for structural change, to increase the local concentration of protein, and to orient protein monomers relative to each other (2, 113). A proteins adopt -helical structure in association with lipid membranes at low lipid-to-protein ratios (315), at high cholesterol concentrations (145), or in conjunction with metal ions (21, 76, 77). Some investigators have found that detergent micelles promote -helical structure (283), whereas others nd that it promotes the formation of oligomers with structure (261). Spontaneous insertion into various membranes has been observed for A segments (82) and for A proteins at relatively low membrane surface pressures (94). The relative abundance of various lipid classes in membranes is altered in AD (121, 227, 247, 252, 350), and altering membrane composition protects PC12 cells from toxic effects of A proteins (338).

Downloaded from physiologyonline.physiology.org on January 24, 2012

Enter Lipid Membranes


Investigations into the role of lipids in AD are experimentally challenging because they are chemically diverse, with thousands of distinct molecular species present in every cell. They are also physically diverse, rarely existing as monomeric species in solution unless protein bound. In vitro, the vast majority form micelles or vesicles, and the latter may be unilamellar or multilamellar. Thus lipid suspensions generally have two or more phases and complex interphase equilibria. This physical heterogeneity complicates most types of physicochemical analysis, even when pure synthetic lipid preparations are used, and it makes some others outright impossible. It also impedes the measurement of bril formation. Turbidity measurements, for example, are confounded by ambiguity over whether the species causing the turbidity is protein or lipid. Lipids markedly increase the uorescence of the thioavin T apart from bril formation, whereas assays based on Congo Red absorbance ratios entail practical restrictions on protein and lipid concentrations that limit the exibility of this assay. The presence of membranes can also complicate sample preparation, and they yield artifacts when the halogenated solvents used to disaggregate A proteins are not completely removed (56, 310). Due in part to these challenges, the evidence that cell membranes are involved in the pathogenesis of AD remains largely circumstantial, even though the amount of evidence pointing to some
56

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
Altered physical properties, presumably arising from compositional differences, have been observed in hippocampal membranes of AD brains (364). Plasmalogen deciency is frequently associated with AD (102, 110 112, 119, 120, 229). In addition to having an effect on membrane physical properties, plasmalogens are particularly susceptible to oxidative damage (156 158). This susceptibility may confer on plasmalogens the ability to protect other lipid species by diverting and trapping oxidizing agents (36, 117, 118, 170, 195, 196, 222, 230, 242, 264, 362). Cerebral white matter is enriched in plasmalogens for unknown reasons (101). Human epidemiological studies support a link between AD and the consumption of -3 polyunsaturated fatty acyl (PUFA) chains (103, 141, 223, 224, 237, 250, 275, 323), and this association is supported by animal model and cell culture studies (5355, 126, 146, 185, 193, 350). The most prevalent -3 PUFA in the brain is docosahexaenoic acid (DHA), and dietary DHA supplementation alleviates both AD-like histopathology and cognitive impairment in animal models (53, 55, 126, 185). The metabolism of DHA in normal brain is remarkable in several respects (159), and it is difcult to induce a measurable deciency of DHA-containing lipids in the brain tissue of animal models through dietary restriction (84). Animal studies have shown that the brain responds to a dietary deciency of DHA by elongating arachidonic acid (ARA) chains. Because there are no enzymes capable of desaturating the distal end of these PUFA chains, the result is a marked increase in docosapentaenoic acid (DPA) chains (138). A proteins appear to have special relationships or interactions with specic lipid species. For example, the ganglioside GM1 is bound together with A proteins in diffuse amyloid plaques (348). GM1containing membranes promote the formation of structure (212, 215), structure (206), or brils in vitro (67 69, 115, 127, 148 151, 165, 207, 218, 239, 240, 334, 335). In several of these reports, GM1 is presented in raft-like membrane subdomains in which cholesterol is a signicant component, and rafts have also been discussed as an inuence on the processing of APP into A proteins (74). Apart from rafts, cholesterol has been epidemiologically (289, 311) and experimentally (263, 300) linked to the incidence of AD, and it may inuence the production (108, 361), location (77, 87, 145), behavior (79, 214, 352), or toxicity (38, 78, 137, 308, 338) of A proteins. In models of Niemann-Pick Type C disease, A proteins accumulate along with cholesterol (347). Cholesterol depletion reduced the -cleavage of APP and the production of A proteins in a study of neurons in the rat hippocampus (288). Treatment with lovastatin/mevalonate alone, however, was insufcient to induce a signicant effect; treatment with -cyclodextrin to achieve a 70% reduction of cellular cholesterol content was also required. It has been suggested that the dependence of A protein production on cholesterol is due to the selective activity of -secretase activity in cholesterol-dependent raft-like subdomains of the plasma membrane, whereas -secretase activity appears to predominate in non-raft domains (95). Some -secretase stimulation has also been attributed to neutral glycosphingolipids and anionic phospholipids (152). The efcacy of a -secretase inhibitor has been increased by linking it to cholesterol and thereby targeting it to membranes (260).

The Membrane as Villain


Downloaded from physiologyonline.physiology.org on January 24, 2012 The lipids in cell membranes are often regarded as being chemically unreactive and merely a physical barrier or a support matrix for proteins. That view is misleading on many levels, of course, but particularly so when the membrane lipids contain PUFA chains and are subjected to oxidative stress. Reviews of the role of membranes in AD pathogenesis frequently overlook the effects of oxidative stress and chemical modication of PUFA chains on membrane properties. A proteins have a prooxidant activity toward polyunsaturated lipids that can be neutralized by lipophilic antioxidants, chelation of metal ions, anaerobic conditions, mutation of His13 or His14 to Ala, or modication of the Met35 side chain (232). Lipid oxidation products and the susceptibility of lipids to oxidative damage are both increased in AD (78, 104, 210, 309). Several reports have outlined the potential for a complex interplay between cholesterol oxidation products and A proteins (38, 116, 235, 241, 301, 329, 356, 360). The myriad mechanisms of oxidative stress yield diverse chemically reactive products from PUFA chains including hydroxy- and hydroperoxy-lipids that undergo spontaneous decomposition, lipid free radicals that may participate in free-radical chain reactions, as well as fragments such as malondialdehyde, acrolein, and hydroxynonenal with the potential to form adducts with proteins and nucleic acids (61, 270, 274, 293, 295, 296). These materials are direct toxic threats to the tissues in which they are generated. Brain is the most lipidrich organ in the body, and it contains more lipids bearing PUFA chains than any other organ. Therefore, brain tissue is at particularly high risk for chemical injury due to highly reactive lipid oxidation products. Evidence that lipid peroxidation may be involved in the pathogenesis of AD has been extensively reviewed (13, 42, 46, 201). Lipid hydroperoxides

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

57

REVIEWS
undergo spontaneous (nonenzymatic) decomposition, and -6 PUFA chains yield reactive , -unsaturated 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 mentioned above can catalyze the formation of HNE from -6 PUFAs in the presence of copper ions (232). Isoprostanes are relatively unreactive compounds that have been used as biomarkers of oxidative stress (179, 221, 244, 268). Some reports suggest that isoprostanes are specically elevated in AD (253, 255, 256), although these ndings have not been conrmed by all investigators (221, 357). Analogous products derived from -3 PUFA chains have been dubbed neuroprostanes and also have been put forward as indexes of oxidative stress in the brain (14, 266, 267, 298). Compared with isoprostanes, HNE is chemically reactive, with a well known propensity to form adducts with the side chains of various amino acid residues (37, 98, 328). For this reason, HNE-protein adducts have also been used as biomarkers of oxidative stress (325). HNE concentrations in human ventricular uid are 15 M and are elevated in AD (190, 203, 210). HNE modication is also known to inhibit proteasome function (286) and glutamate transport in synaptosomes (177). Together with the observation that immunoreactivity of antibodies to HNE-modied His residues localizes to amyloid plaques (7, 294), these observations suggest that A proteins not only promote lipid oxidation but that there may also be a mechanistic link between the lipid oxidation products formed during oxidative stress and A misfolding (161). A proteins increase HNE production from PUFA chains in vitro that, in turn, causes covalent modication of the three His residues in A proteins and bril formation (231). These modications promote the aggregation of A proteins into brils (FIGURE 2) (165, 231, 232). Moreover, they increase the ability of A 42 to seed bril formation by unmodied A 40 (166). The effects of HNE on A bril formation depend on the size or hydrophobicity of the modication because the corresponding analog from -3 PUFA chains, 4-hydroxy-2(E)-hexenal, does not have this effect (187). The addition of an octanoyl group to specic Lys residues of A proteins also appears to have a pro-amyloidogenic effect (258). In vivo, HNE-His epitopes are concentrated in the vicinity of amyloid plaques, but they do not precisely co-localize with the plaques (FIGURE 3). One possible explanation for this distribution is that the presence of HNE-His adducts and Cu-His complexes in the same protein molecule are mutually exclusive. Presumably, the formation of CuHis complexes precedes the generation of HNE and HNE-His adducts, if indeed they are responsi58

ble for generating HNE from -6 PUFAs. Consequently, HNE-His epitopes would develop around preexisting amyloid plaques, not within them. A second possibility is that A -specic antibody epitopes are masked by HNE modication, as observed in vitro for 4G8 and 6E10 epitopes (231). A third possibility is that A brils induced to form by HNE modication have relatively sparse HNEHis epitopes (as suggested by FIGURE 2), yielding sparse uorescence among brils. The uorescence of HNE-11S seen in FIGURE 3 may be to HNE produced in the plaque but bound to HNEHis adducts in proteins other than A proteins. Some investigators have focused on the role of acrolein in the pathogenesis of AD. Acrolein is a well known environmental toxin and a recently recognized product of lipid peroxidation (324, 326, 327). It reacts spontaneously with Lys residue side chains, forming a cyclic N-(3-formyl-3,4-dehydropiperidino) derivative that has been considered a biomarker for measuring oxidative stress in AD (52). Acrolein is elevated in AD brain, it appears to be more neurotoxic than HNE, it causes elevated intracellular calcium levels (192, 342), and it appears to inactivate ippase, inducing a breakdown of lipid membrane asymmetry and apoptosis (1, 59).

Downloaded from physiologyonline.physiology.org on January 24, 2012

The Membrane as Victim


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

FIGURE 2. Electron micrograph of brils induced by HNE modication


A 42 was induced to brillize with HNE, then treated with mouse anti-His-HNE, and gold-tagged anti-mouse antibody. The focus in this image is on the gold particles, but various bril morphologies are evident, and the gold particles making anti-HNE-His antibodies are preferentially associated with long, relatively straight bils.

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
physical attacks on the barrier function of membranes (265). For example, A proteins or their fragments have been observed to penetrate the membrane surface (82, 94), disrupt raft-like domains (67), cross the membrane (341), induce isotropic phases (176), increase the permeability of membranes to various materials (154, 344), cause physical thinning (81), decrease the mobility of hydrophobic membrane probes (169), activate phospholipase A2 (182), and promote membrane fusion (81, 249). Membrane binding by A proteins appears to mediate some forms of neurotoxicity (21, 70, 316). Plasma membranes isolated from human brain accelerate A brillogenesis (339), whereas brillizing A proteins disrupt the structure of membranes formed from both synthetic lipids (168, 169) and whole-brain lipid extracts (353). The action of A proteins on membranes has been compared with the action of antimicrobial peptides, and homogenates of brain with AD have elevated antimicrobial activity (299). Among the effects of A proteins on membranes, however, the formation of ion channels has received the most attention (10 12, 93, 147, 186, 251). A proteins share an ability to induce channel-like activity in membranes with a wide variety of other amyloid-forming proteins (259). In the case of A , the channels exhibit only modest cation selectivity but very long lifetimes. A cation channels can be blocked by Zn2 , suggesting the possibility that they may be therapeutic targets (9), and small molecule inhibitors of the A calcium channel have been described (88). Ring-like structures possibly representing these pores or channels have been observed in membranes by AFM (173, 186, 259) and EM (174, 175). Although such features are not always observed with A proteins (114, 341), there are many reports suggesting that they are formed by other amyloidogenic proteins such as the insulin-associated polypeptide, -synuclein, and serum amyloid A protein (259). It has been suggested that the channel-forming toxic properties of A proteins on a membrane depends on the extent to which it has aggregated into oligomers and that this extent is concentration dependent (276). Model structures of the purported channels have been patterned after -barrel poreforming toxins (8, 93). This comparison is suggested not only by the EM/AFM images but by the reactivity of A and -hemolysin with so-called conformation-specic antibodies (354). Recent studies conrming the formation of zinc-sensitive ion channels have also exposed the potential for artifact due to residual amounts of halogenated solvent in the A protein samples (56). It may be signicant that truncated A peptides that are unable to form brils can nevertheless induce ion permeability (142) and that membranes formed from highly compressible lipids are most sensitive to A -induced changes in permeability (297). It has been suggested that channel openings are merely protein-induced defects in the bilayer structure or organization that heal after a time, giving the appearance of a transient channel opening (114). This mechanism would be consistent with oligomer-induced membrane thinning (297) and would help resolve the paradoxical appearance of many simultaneous pore structures in membranes that exhibit only single channel conductances (96). In living cells, as opposed to synthetic systems, one must consider the possibility that A may profoundly affect the behavior of naturally occurring channels and transporters. For example, A 42 increases HNE-modication of a glutamate transporter in synaptosomes (177). A oligomers have been implicated in causing a pathological calcium inux through hippocampal NMDA receptors, followed by the activation of calpain and the degradation of dynamic 1-a GTPase involved in synaptic vesicle recycling (155). A proteins have also been shown to depress the surface concentration of AMPA receptors involved in calcium regulation at

Downloaded from physiologyonline.physiology.org on January 24, 2012

FIGURE 3. Immunouorescent staining of the posterior parietal association area in a transgenic mouse model of Alzheimers disease that expressed amyloid precursor protein with the Swedish K670N/M671L mutation, and presenilin 1 with the M146L mutation
A: 2H4 antibodies (Covance) specic for the amino terminus A proteins (green). B: HNE 11S specic for HNE-His adducts (red). C: A and B superimposed. Possible reasons for the close association of HNE-His epitopes and A proteins, but not precise colocalization, are discussed in the text.

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

59

REVIEWS
the synapse (133, 188). The dysregulation of calcium has long been associated with AD pathogenesis (172) and is thought to be involved with the accumulation of amyloid deposits (85, 208, 209) and the hyperphosphorylation of tau (25). Calcium also promotes the conversion of oligomeric A to brils (140). An extensive literature exists on the role of mitochondria in AD pathogenesis, and the accumulation of calcium by mitochondria followed by damage to the mitochondrial membrane structure is a prominent theme in this literature (131, 246, 262). copies confer an eightfold increased risk. One copy of the ApoE2 gene, however, reduces the risk by 60% (73). In addition to the epidemiological evidence, there is an abundance of experimental evidence linking ApoE to the pathogenesis of Alzheimers disease. For example, the ApoE4 allele is associated with increased A deposits in the brain, and a distinct neuropathological phenotype (278), whereas a lack of ApoE reduces A deposition in mice (19). Amyloid plaques bind anti-ApoE antibodies, suggesting that ApoE is present in these plaques (3). Indeed, ApoE copuries with A from amyloid plaques (26) and may exist as a bound complex with A proteins (245, 349). Some have suggested that ApoE4 accelerates bril formation by A 40 (18, 194, 271, 343), but its behavior in this regard may depend on whether it is monomeric or dimeric. Others have suggested that dimeric forms inhibit bril formation and that ApoE3 is much more effective at doing this because a large fraction circulates as a disulde-linked dimer, whereas ApoE4 cannot form disulde bonded dimers due to its C112R substitution (99, 340). Despite all of these observations, the reason that different isoforms of ApoE have different levels of risk for AD is not known. There are no isoform-dependent differences in the apparent structure of lipoprotein particles (280). A case for isoform-specic direct interactions between ApoE and A proteins has been made (233), but clear conclusions are elusive due to problems with protein purity, aggregation and denaturation of the ApoE protein, and indirect assay methods (4, 171, 198, 245, 303, 307). Others have suggested that risk of AD with ApoE isoforms may be related to differences in antioxidant activity (178, 217, 312). ApoE4 has no Cys residues and, hence, no free thiol groups; ApoE3 has one Cys, whereas ApoE2 has two. Free thiol groups have signicant antioxidant activity via mechanisms that differ from those of glutathione (109, 333, 359). In apoA-I, variants with a free Cys residue side chain exhibit signicantly greater antioxidant activity than variants without a free Cys residue (29). Therefore, it is intriguing to speculate that ApoE4 confers increased risk of AD, whereas ApoE2 confers decreased risk, because Cys residue side chains protect lipids in lipoprotein E particles against oxidative stress.

The Lipids Associated With Apolipoprotein E


The lipids in nascent lipoprotein particles are arranged in a membrane-like bilayer, surrounded by apolipoprotein molecules in a conguration often called the belt model (FIGURE 4) (280). Apolipoprotein E (ApoE) is a focus of considerable interest because its isoforms are strongly associated with the risk of developing AD. In plasma, ApoE is found with other proteins in lipoprotein particles as diverse as chylomicrons and high-density lipoproteins. In the brain and cerebrospinal uid, ApoE is the most abundant apolipoprotein and is found in particles that resemble high-density lipoproteins in both density and size (269). Among the three common isoforms, ApoE3 is the most common (77% of the alleles) and is therefore considered to be the wild type. ApoE2 has an R158C substitution, whereas ApoE4 has a C112R substitution, and both are associated with forms of hyperlipidemia (197). One copy of the ApoE4 gene confers a threefold increased risk of Alzheimers disease, whereas two

Downloaded from physiologyonline.physiology.org on January 24, 2012

FIGURE 4. Nascent lipoprotein E particles consist of -helical belts around the perimeter of a lipid bilayer disk
For this arrangement and the number of protein and lipid molecules in a particle, there is excess protein, which may situate across the face of the disk. When the protein is apoE3, there is one thiol group for every 45 lipids, concentrating a large antioxidant capacity in close proximity to the lipids. 60

Toward a Comprehensive Theory of AD Pathogenesis


The cause of AD is known in cases of familial disease, but that knowledge has not claried how sporadic disease develops, nor has it so far yielded an effective therapy for either form of the disease. Clearly, we need a more detailed theory of AD

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
pathogenesis, and it seems likely that lipid membranes will have a prominent role in any such theory. As summarized above, the interactions between lipid membranes and A proteins are bidirectional: lipids damage A proteins, and A proteins damage lipid membranes. These processes can operate in tandem to accelerate bril formation in human brain lipid extracts (231). Even if bril formation is only an inert by-product of the disease-causing process, it is nonetheless a pathognomonic nding of AD. Therefore, it is vital to understand how brils form. As illustrated in FIGURE 5, the interaction of lipid membranes may constitute an amplication system for bril formation. A proteins in vitro only form brils at micromolar concentrations and after days of incubation, whereas membranes containing PUFA-chains lower the protein concentration requirement by three orders of magnitude and shorten the time required to minutes (165, 166). The same system may also amplify multiple mechanisms of neurotoxicity that operate independently of bril formation and link many of the seemingly unrelated observations reviewed above. For example, inammation due to trauma or other factors will promote oxidative stress and the production of reactive oxygen species that exert direct neurotoxicity or lipid damage and A protein aggregation. Lipid oxidation products may exert direct toxicity. ApoE4 proteins are decient in thiol-mediated antioxidant activity; this deciency would allow excess oxidative damage to the lipids in lipoprotein particles that likewise promote A protein aggregation. Aging is associated with reduced tissue antioxidant levels, accumulated oxidative lipid damage, and low levels of adventitial A protein aggregation, with each process promoting further A protein aggregation. Various aggregated forms of A protein may form pores, channels, or other neurotoxic structures in neuronal membranes. It is not yet clear whether all cases of AD arise through the operation of one primary pathogenic mechanism that diverges only at a late stage into various subtypes or whether disparate mechanisms operate from the beginning. For example, neurobrillary tangles of tau protein (20, 322) occur in only 70 80% of cases of AD (122), suggesting that tangles reect a response to some but not all pathogenic paths. In any case, one or more additional factors must be incorporated into the amyloid hypothesis to explain the pathogenesis of AD, and confronting the experimental challenges presented by lipids and membranes may be necessary to identify such factors.
No conicts of interest, nancial or otherwise, are declared by the author(s).

FIGURE 5. Membrane-mediated amplication of brillogenesis, illustrating possible relationships between meachanisms involved in A protein aggregation and neurotoxic mechanisms involved in the pathogenesis of Alzheimers disease
Lipid oxidation products modify the three His residues of A 42, increasing its membrane afnity and accelerating the conversion of A 40 into oligomers and brils. Oligomeric A proteins bind copper ions while they undergo redox cycling. Highly reactive oxygen species may be generated by electrons from copper or as by-products of inammation, including inammation induced by trauma. ApoE4 alleles lack thiol-mediated antioxidant activity and may allow excess oxidative damage to the lipids in lipoprotein particles. Aging by itself is associated with reduced tissue antioxidant levels and accumulated oxidative lipid 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 in neuronal membranes. The authors are supported by grants from the National Institute on Aging, the Alzheimers Association, and the American Health Assistance Foundation.

Downloaded from physiologyonline.physiology.org on January 24, 2012

References
1. Abdul HM, Buttereld DA. Protection against amyloid betapeptide (1-42)-induced loss of phospholipid asymmetry in synaptosomal membranes by tricyclodecan-9-xanthogenate (D609) and ferulic acid ethyl ester: implications for Alzheimers disease. BBA Mol Basis Dis 1741: 140 148, 2005. Aisenbrey C, Borowik T, Bystrom R, Bokvist M, Lindstrom F, Misiak H, Sani MA, Grobner G. How is protein aggregation in amyloidogenic diseases modulated by biological membranes? Eur Biophys J Biophys Lett 37: 247255, 2008. Aizawa Y, Fukatsu R, Takamaru Y, Tsuzuki K, Chiba H, Kobayashi K, Fujii N, Takahata N. Amino-terminus truncated apolipoprotein E is the major species in amyloid deposits in Alzheimers disease affected brains: a possible role for apolipoprotein E in Alzheimers disease. Brain Res 768: 208 214, 1997. Aleshkov S, Abraham CR, Zannis VI. Interaction of nascent ApoE2, ApoE3, and ApoE4 isoforms expressed in mammalian cells with amyloid peptide beta (1 40). Relevance to Alzheimers disease. Biochemistry 36: 1057110580, 1997. Ali FE, Separovic F, Barrow CJ, Cherny RA, Fraser F, Bush AI, Masters CL, Barnham KJ. Methionine regulates copper/hydrogen peroxide oxidation products of A beta. J Pept Sci 11: 353360, 2005. Ali FE, Separovic F, Barrow CJ, Yao SG, Barnham KJ. Copper and zinc mediated oligomerisation of A beta peptides. Int J Pept Res Ther 12: 153164, 2006. Ando Y, Brannstrom T, Uchida K, Nyhlin N, Nasman B, Suhr O, Yamashita T, Olsson T, El Salhy M, Uchino M, Ando M. Histochemical detection of 4-hydroxynonenal protein in Alzheimer amyloid. J Neurol Sci 156: 172176, 1998. Arispe N. Architecture of the Alzheimers A beta P ion channel pore. J Membrane Biol 197: 33 48, 2004.

2.

3.

4.

5.

6.

7.

8.

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

61

REVIEWS
9. Arispe N, Diaz JC, Simakova OA. Ion channels. Prospects for treating Alzheimers disease with A channel blockers. BBA Biomembranes 1768: 19521965, 2007. 25. Baudier J, Cole RD. Phosphorylation of tau-proteins to a state like that in Alzheimers brain is catalyzed by a calcium calmodulin-dependent kinase and modulated by phospholipids. J Biol Chem 262: 1757717583, 1987. 26. Baumann MH, Kallijarvi J, Lankinen H, Soto C, Haltia M. Apolipoprotein E includes a binding site which is recognized by several amyloidogenic polypeptides. Biochem J 349: 77 84, 2000. 27. Behl C, Davis JB, Lesley R, Schubert D. Hydrogen peroxide mediates amyloid beta protein toxicity. Cell 77: 817 827, 1994. 28. Berthon G. Does human beta A4 exert a protective function against oxidative stress in Alzheimers disease? Med Hypotheses 54: 672 677, 2000. 29. Bielicki JK, Oda MN. Apolipoprotein A-I-Milano and apolipoprotein A-I-Paris exhibit an antioxidant activity distinct from that of wild-type apolipoprotein A-I. Biochemistry 41: 2089 2096, 2002. 30. Bieschke J, Zhang Q, Powers ET, Lerner RA, Kelly JW. Oxidative metabolites accelerate Alzheimers amyloidogenesis by a two-step mechanism, eliminating the requirement for nucleation. Biochemistry 44: 4977 4983, 2005. 31. Bishop GM, Robinson SR. The amyloid paradox: amyloid-beta-metal complexes can be neurotoxic and neuroprotective. Brain Pathol 14: 448 452, 2004. 32. Bitan G, Tarus B, Vollers SS, Lashuel HA, Condron MM, Straub JE, Teplow DB. A molecular switch in amyloid assembly: Met(35) and amyloid beta-protein oligomerization. J Am Chem Soc 125: 15359 15365, 2003. 33. Bokvist M, Lindstrom F, Watts A, Grobner G. Two types of Alzheimers beta-amyloid (1-40) peptide membrane interactions: aggregation preventing transmembrane anchoring versus accelerated surface bril formation. J Mol Biol 335: 1039 1049, 2004. 34. Boyd-Kimball D, Castegna A, Sultana R, Poon HF, Petroze R, Lynn BC, Klein JB, Buttereld DA. Proteomic identication of proteins oxidized by A beta(1-42) in synaptosomes: implications for Alzheimers disease. Brain Res 1044: 206 215, 2005. 35. Brezesinski G, Maltseva E, Mohwald H. Adsorption of amyloid beta (1-40) peptide at liquid interfaces. Z Phys Chem 221: 95111, 2007. 36. Brosche T, Platt D. Mini-review: The biological signicance of plasmalogens in defense against oxidative damage. Exp Gerontol 33: 363369, 1998. 37. Bruenner BA, Jones AD, German JB. Direct characterization of protein adducts of the lipid-peroxidation product 4-hydroxy-2-nonenal using electrospray mass-spectrometry. Chem Res Tox 8: 552559, 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)-hydroxycholesterol content in the brain and ameliorates amyloid pathology in mice with AD. Proc Natl Acad Sci USA 107: 30813086, 2010. 39. Bush AI. The metallobiology of Alzheimers disease. Trends Neurosci 26: 207214, 2003. 40. Buttereld DA. Amyloid beta-peptide (1-42)-induced oxidative stress and neurotoxicity: implications for neurodegeneration in Alzheimers disease brain. A review. Free Radic Res 36: 1307 1313, 2002. 41. Buttereld DA. Proteomics: a new approach to investigate oxidative stress in Alzheimers disease brain. Brain Res 1000: 17, 2004. 42. Buttereld DA, Boyd-Kimball D. Amyloid betapeptide(1-42) contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol 14: 426 432, 2004. 43. Buttereld DA, Boyd-Kimball D. The critical role of methionine 35 in Alzheimers amyloid-betapeptide (1-42)-induced oxidative stress and neurotoxicity. Biochim Biophys Acta 1703: 149 156, 2005. 44. Buttereld DA, Boyd-Kimball D, Castegna A. Proteomics in Alzheimers disease: insights into potential mechanisms of neurodegeneration. J Neurochem 86: 13131327, 2003. 45. Buttereld DA, Bush AI. Alzheimers amyloid beta-peptide (1-42): involvement of methionine residue 35 in the oxidative stress and neurotoxicity properties of this peptide. Neurobiol Aging 25: 563568, 2004. 46. Buttereld DA, Castegna A, Lauderback CM, Drake J. Evidence that amyloid beta-peptide-induced lipid peroxidation and its sequelae in Alzheimers disease brain contribute to neuronal death. Neurobiol Aging 23: 655 664, 2002. 47. Buttereld DA, Galvan V, Lange MB, Tang HD, Sowell RA, Spilman P, Fombonne J, Gorostiza O, Zhang JL, Sultana R, Bredesen DE. In vivo oxidative stress in brain of Alzheimer disease transgenic mice: requirement for methionine 35 in amyloid beta-peptide of APP. Free Radic Biol Med 48: 136 144, 2010. 48. Buttereld DA, Hensley K, Harris M, Mattson MP, Carney JM. Beta-amyloid peptide free-radical fragments initiate synaptosomal lipoperoxidation in a sequence-specic fashion: implications to Alzheimers-disease. Biochem Biophys Res Commun 200: 710 715, 1994. 49. Buttereld DA, Kanski J. Methionine residue 35 is critical for the oxidative stress and neurotoxic properties of Alzheimers amyloid beta-peptide 1-42. Peptides 23: 1299 1309, 2002. 50. Buttereld DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimers disease brain: potential causes and consequences involving amyloid beta-peptide-associated free radical oxidative stress. Free Radic Biol Med 32: 1050 1060, 2002. 51. Buttereld DA, Reed T, Newman SF, Sultana R. Roles of amyloid beta-peptide-associated oxidative stress and brain protein modications in the pathogenesis of Alzheimers disease and mild cognitive impairment. Free Radic Biol Med 43: 658 677, 2007. 52. Calingasan NY, Uchida K, Gibson GE. Proteinbound acrolein: a novel marker of oxidative stress in Alzheimers disease. J Neurochem 72: 751756, 1999. 53. Calon F, Cole G. Neuroprotective action of omega-3 polyunsaturated fatty acids against neurodegenerative diseases: evidence from animal studies. Prost Leuk Essen Fatty Acids 77: 287293, 2007. 54. Calon F, Lim GP, Morihara T, Yang FS, Ubeda O, Salem N, Frautschy SA, Cole GM. Dietary n-3 polyunsaturated fatty acid depletion activates caspases and decreases NMDA receptors in the brain of a transgenic mouse model of Alzheimers disease. 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, Ashe KH, Frautschy SA, Cole GM. Docosahexaenoic acid protects from dendritic pathology in an Alzheimers disease mouse model. Neuron 43: 633 645, 2004. 56. Capone R, Quiroz FG, Prangkio P, Saluja I, Sauer AM, Bautista MR, Turner RS, Yang J, Mayer M. Amyloid-beta-induced ion ux in articial lipid bilayers and neuronal cells: resolving a controversy. Neurotox Res 16: 113, 2009.

10. Arispe N, Pollard HB, Rojas E. Giant multilevel cation channels formed by Alzheimer-disease amyloid beta-protein [A-Beta-P-(1-40)] in bilayermembranes. Proc Natl Acad Sci USA 90: 10573 10577, 1993. 11. Arispe N, Pollard HB, Rojas E. Zn2 interaction with Alzheimer amyloid beta protein calcium channels. Proc Natl Acad Sci USA 93: 1710 1715, 1996. 12. Arispe N, Rojas E, Pollard HB. Alzheimer-disease amyloid beta-protein forms calcium channels in bilayer-membranes: blockade by tromethamine and aluminum. Proc Natl Acad Sci USA 90: 567 571, 1993. 13. Arlt S, Beisiegel U, Kontush A. Lipid peroxidation in neurodegeneration: new insights into Alzheimers disease. Curr Opin Lipid 13: 289 294, 2002. 14. Arneson KO, Roberts LJ. Measurement of products of docosahexaenoic acid peroxidation, neuroprostanes, and neurofurans. Lipidomics Bioactive Lipids 433: 127143, 2007. 15. Atwood CS, Huang XD, Khatri A, Scarpa RC, Kim YS, Moir RD, Tanzi RE, Roher AE, Bush AI. Copper catalyzed oxidation of alzheimer A beta. Cell Mol Biol 46: 777783, 2000. 16. Atwood CS, Obrenovich ME, Liu TB, Chan H, Perry G, Smith MA, Martins RN. Amyloid-beta: a chameleon walking in two worlds: a review of the trophic and toxic properties of amyloid-beta. Brain Res Rev 43: 116, 2003. 17. Atwood CS, Perry G, Zeng H, Kato Y, Jones WD, Ling KQ, Huang XD, Moir RD, Wang DD, Sayre LM, Smith MA, Chen SG, Bush AI. Copper mediates dityrosine cross-linking of Alzheimers amyloid-beta. Biochemistry 43: 560 568, 2004. 18. Bales KR, Verina T, Cummins DJ, Du YS, Dodel TC, Saura J, Fishman CE, DeLong CA, Piccardo P, Petegnief V, Ghetti B, Paul SM. Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimers disease. Proc Natl Acad Sci USA 96: 1523315238, 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. Lack of apolipoprotein E dramatically reduces amyloid beta- peptide deposition. Nat Gen 17: 263264, 1997. 20. Ballatore C, Lee VMY, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimers disease and 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, Barrow CJ, Wade JD, Masters CL, Cherny RA, Curtain CC, Bush AI, Cappai R. Neurotoxic, redox-competent Alzheimers beta-amyloid is released from lipid membrane by methionine oxidation. J Biol Chem 278: 42959 42965, 2003. 22. Barnham KJ, Haeffner F, Ciccotosto GD, Curtain CC, Tew D, Beyreuther K, Carrington D, Masters CL, Cherny RA, Cappai R, Bush AI. Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimers disease -amyloid. FASEB J 18: 14271429, 2004. 23. Baruch-Suchodolsky R, Fischer B. Soluble amyloid beta(1-28)-copper(I)/copper(II)/iron(II) complexes are potent antioxidants in cell-free systems. Biochemistry 47: 7796 7806, 2008. 24. Baruch-Suchodolsky R, Fischer B. A 40, either soluble or aggregated, is a remarkably potent antioxidant in cell-free oxidative systems. Biochemistry 48: 4354 4370, 2009.

Downloaded from physiologyonline.physiology.org on January 24, 2012

62

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
57. Castegna A, Aksenov M, Aksenova M, Thongboonkerd V, Klein JB, Pierce WM, Booze R, Markesbery WR, Buttereld DA. Proteomic identication of oxidatively modied proteins in Alzheimers disease brain. Part 1: creatine kinase bb, glutamine synthase, and ubiquitin carboxy-terminal 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, Buttereld DA. Proteomic identication of oxidatively modied proteins in Alzheimers disease brain. Part II: dihydropyrimidinase-related protein 2, alpha-enolase and heat shock cognate 71. J Neurochem 82: 1524 1532, 2002. 59. Castegna A, Lauderback CM, Mohmmad-Abdul H, Buttereld DA. Modulation of phospholipid asymmetry in synaptosomal membranes by the lipid peroxidation products, 4-hydroxynonenal and acrolein: implications for Alzheimers disease. Brain Res 1004: 193197, 2004. 60. Castegna A, Thongboonkerd V, Klein JB, Lynn B, Markesbery WR, Buttereld DA. Proteomic identication of nitrated proteins in Alzheimers disease brain. J Neurochem 85: 1394 1401, 2003. 61. Castellani RJ, Perry G, Harris PLR, Cohen ML, Sayre LM, Salomon RG, Smith MA. Advanced lipid peroxidation end-products in Alexanders disease. Brain Res 787: 1518, 1998. 62. Caughey B, Lansbury PT. Protobrils, pores, brils, and neurodegeneration: separating the responsible protein aggregates from the innocent bystanders. Ann Rev Neurosci 26: 267298, 2003. 63. Chauhan A, Ray I, Chauhan VPS. Interaction of amyloid beta-protein with anionic phospholipids: possible involvement of Lys(28) and C-terminus aliphatic 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, Moir RD, Lim JT, Beyreuther K, Zheng H, Tanzi RE, Masters CL, Bush AI. Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimers disease transgenic 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 Alzheimers disease A beta amyloid deposits by biometal depletion. J Biol Chem 274: 2322323228, 1999. 66. Chi EY, Ege C, Winans A, Majewski J, Wu GH, Kjaer K, Lee KYC. Lipid membrane templates the ordering and induces the brillogenesis of Alzheimers disease amyloid-beta peptide. Proteins 72: 124, 2008. 67. Chi EY, Frey SL, Lee KYC. Ganglioside GM1mediated amyloid-beta brillogenesis and membrane disruption. Biochemistry 46: 19131924, 2007. 68. Choo-Smith LP, Garzon-Rodriguez W, Glabe CG, Surewicz WK. Acceleration of amyloid bril formation by specic binding of Abeta-(1-40) peptide to ganglioside-containing membrane vesicles. J Biol Chem 272: 2298722990, 1997. 69. Choo-Smith LP, Surewicz WK. The interaction between Alzheimer amyloid beta(1-40) peptide and ganglioside GM1-containing membranes. FEBS Lett 402: 9598, 1997. 70. Ciccotosto GD, Tew D, Curtain CC, smith D, Carrington D, Masters CL, Bush AI, Cherny RA, Cappai R, Barnham KJ. Enhanced toxicity and cellular binding of a modied amyloid beta peptide with a methionine to valine substitution. J Biol Chem 279: 28425 42538, 2004. 71. Cirrito JR, Yamada KA, Finn MB, Sloviter RS, Bales KR, May PC, Schoepp DD, Paul SM, Mennerick S, Holtzman DM. Synaptic activity regulates interstitial uid amyloid-beta levels in vivo. Neuron 48: 913922, 2005. 72. Cleary JP, Walsh DM, Hofmeister JJ, Shankar GM, Kuskowski MA, Selkoe DJ, Ashe KH. Natural oligomers of the amyloid-protein specically disrupt cognitive function. Nat Neurosci 8: 79 84, 2005. 73. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericakvance MA. Gene dose of apolipoprotein-E type-4 allele and the risk of Alzheimers-disease in late-onset families. Science 261: 921923, 1993. 74. Cordy JM, Hooper NM, Turner AJ. The involvement of lipid rafts in Alzheimers disease. Mol Membr Biol 23: 111122, 2006. 75. Cuajungco MP, Goldstein LE, Nunomura A, Smith MA, Lim JT, Atwood CS, Huang X, Farrag YW, Perry G, Bush AI. Evidence that the -amyloid plaques of Alzheimers disease represent the redox-silencing and entombment of abeta by zinc. 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. Alzheimers eisease amyloid-beta binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits. J Biol Chem 276: 20466 20473, 2001. 77. Curtain CC, Ali FE, Smith DG, Bush AI, Masters CL, Barnham KJ. Metal ions, pH, and cholesterol regulate the interactions of Alzheimers disease amyloid-beta peptide with membrane lipid. J Biol Chem 278: 29772982, 2003. 78. Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatanpaa K, Troncoso JC, Mattson MP. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimers disease. Proc Natl Acad Sci USA 101: 2070 2075, 2004. 79. DErrico G, Vitiello G, Ortona O, Tedeschi A, Ramunno A, DUrsi AM. Interaction between Alzheimers A beta(25-35) peptide and phospholipid bilayers: the role of cholesterol. Biochim Biophys Acta 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 protein oxidation to relevant signs of severe functional impairment. Free Radic Biol Med 31: 1075 1083, 2001. 81. Dante S, Hauss T, Brandt A, Dencher NA. Membrane fusogenic activity of the Alzheimers peptide A beta(1-42) demonstrated by small-angle neutron scattering. J Mol Biol 376: 393 404, 2008. 82. Dante S, Hauss T, Dencher NA. Insertion of externally administered amyloid beta peptide 25-35 and perturbation of lipid bilayers. Biochemistry 42: 1366713672, 2003. 83. de Planque MRR, Raussens V, Contera SA, Rijkers DTS, Liskamp RMJ, Ruysschaert JM, Ryan JF, Separovic F, Watts A. beta-sheet structured beta-amyloid(1-40) perturbs phosphatidylcholine model membranes. J Mol Biol 368: 982997, 2007. 84. DeMar JC, Ma KZ, Bell JM, Rapoport SI. Halflives of docosahexaenoic acid in rat brain phospholipids are prolonged by 15 weeks of nutritional deprivation of n-3 polyunsaturated fatty acids. J Neurochem 91: 11251137, 2004. 85. Demuro A, Parker I, Stutzmann GE. Calcium signaling and amyloid toxicity in Alzheimers disease. J Biol Chem 285: 1246312468, 2010. 86. Deshpande A, Kawai H, Metherate R, Glabe CG, Busciglio J. A role for synaptic zinc in activitydependent A beta oligomer formation and accumulation at excitatory synapses. J Neurosci 29: 4004 4015, 2009. 87. Devanathan S, Salamon Z, Lindblom G, Grobner G, Tollin G. Effects of sphingomyelin, cholesterol and zinc ions on the binding, insertion and aggregation of the amyloid A beta(1-40) peptide in solid-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 Alzheimer A beta calcium channel potently protect neurons from A beta cytotoxicity. Proc Natl Acad Sci USA 106: 3348 3353, 2009. 89. Dikalov SI, Vitek MP, Maples KR, Mason RP. Amyloid beta peptides do not form peptide-derived free radicals spontaneously, but can enhance metal-catalyzed oxidation of hydroxylamines to nitroxides. J Biol Chem 274: 93929399, 1999. 90. Donnelly PS, Xiao ZG, Wedd AG. Copper and Alzheimers disease. Curr Opin Chem Biol 11: 128 133, 2007. 91. Drake J, Link CD, Buttereld DA. Oxidative stress precedes brillar deposition of Alzheimers disease amyloid beta-peptide (1-42) in a transgenic Caenorhabditis elegans model. Neurobiol Aging 24: 415 420, 2003. 92. Drew SC, Noble CJ, Masters CL, Hanson GR, Barnham KJ. Pleomorphic copper coordination by Alzheimers disease amyloid-beta peptide. J Am Chem Soc 131: 11951207, 2009. 93. Durell SR, Guy HR, Arispe N, Rojas E, Pollard HB. Theoretical-models of the ion-channel structure of amyloid beta-protein. Biophys J 67: 2137 2145, 1994. 94. Ege C, Lee KYC. Insertion of Alzheimers A beta 40 peptide into lipid monolayers. Biophys J 87: 17321740, 2004. 95. Ehehalt R, Keller P, Haass C, Thiele C, Simons K. Amyloidogenic processing of the Alzheimer beta-amyloid precursor protein depends on lipid rafts. J Cell Biol 160: 113123, 2003. 96. Eliezer D. Amyloid ion channels: a porous argument 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 mechanism of formation of 4-hydroxynonenal during microsomal lipid-peroxidation. Biochim Biophys Acta 876: 154 166, 1986. 98. Esterbauer H, Schaur RJ, Zollner H. Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11: 81128, 1991. 99. Evans KC, Berger EP, Cho CG, Weisgraber KH, Lansbury PT. Apolipoprotein E is a kinetic but not a thermodynamic inhibitor of amyloid formation: implications for the pathogenesis and treatment of Alzheimer disease. Proc Natl Acad Sci USA 92: 763767, 1995. 100. Faller P, Hureau C. Bioinorganic chemistry of copper and zinc ions coordinated to amyloidbeta peptide. Dalton Trans 21: 1080 1094, 2009. 101. Farooqui AA, Horrocks LA. Plasmalogens: Workhorse lipids of membranes in normal and injured neurons and glia. Neuroscientist 7: 232245, 2001. 102. Farooqui AA, Rapoport SI, Horrocks LA. Membrane phospholipid alterations in Alzheimers disease: Deciency of ethanolamine plasmalogens. Neurochem Res 22: 523527, 1997. 103. Friedland RP. Fish consumption and the risk of Alzheimer disease - Is it time to make dietary recommendations? Arch Neurol 60: 923924, 2003.

Downloaded from physiologyonline.physiology.org on January 24, 2012

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

63

REVIEWS
104. Galbusera C, Facheris M, Magni F, Galimberti G, Sala G, Tremolada L, Isella V, Guerini FR, Appollonio I, Galli-Kienle M, Ferrarese C. Increased susceptibility to plasma lipid peroxidation in Alzheimer disease patients. Curr Alzheimer Res 1: 103109, 2004. 105. Garzon-Rodriguez W, Yatsimirsky AK, Glabe CG. Binding of Zn(II), Cu(II), and Fe(II) ions to Alzheimers A beta peptide studied by uorescence. Bioorg Med Chem Lett 9: 22432248, 1999. 106. Gehman JD, OBrien CC, Shabanpoor F, Wade JD, Separovic F. Metal effects on the membrane interactions of amyloid-beta peptides. Eur Biophys J Biophys Lett 37: 333344, 2008. 107. Ghezzi P, Bonetto V. Redox proteomics: Identication of oxidatively, modied proteins. Proteomics 3: 11451153, 2003. 108. Ghribi O, Larsen B, Schrag M, Herman MM. High cholesterol content in neurons increases BACE, beta-amyloid, and phosphorylated tau levels in rabbit hippocampus. Exp Neurol In Press: 2006. 109. Giles GI, Jacob C. Reactive sulfur species: an emerging concept in oxidative stress. Biol Chem 383: 375388, 2002. 110. Ginsberg L, Raque S, Xuereb JH, Rapoport SI, Gershfeld NL. Disease and anatomic specicity of ethanolamine plasmalogen deciency in Alzheimers-disease brain. Brain Res 698: 223226, 1995. 111. Ginsberg L, Xuereb JH, Gershfeld NL. Membrane instability, plasmalogen content, and Alzheimers disease. J Neurochem 70: 25332538, 1998. 112. Goodenowe DB, Cook LL, Liu J, Lu Y, Jayasinghe DA, Ahiahonu PWK, Heath D, Yamazaki Y, Flax J, Krenitsky KF, Sparks DL, Lerner A, Friedland RP, Kudo T, Kamino K, Morihara T, Takeda M, Wood PL. Peripheral ethanolamine plasmalogen deciency: a logical causative factor in Alzheimers disease and dementia. J Lipid Res 48: 24852498, 2007. 113. Gorbenko GP, Kinnunen PKJ. The role of lipidprotein interactions in amyloid-type protein bril formation. 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 within mica supported lipid bilayers induced by the amyloidogenic human amylin peptide. J Mol Biol 342: 877 887, 2004. 115. Gylys KH, Fein JA, Yang F, Miller CA, Cole GM. Increased cholesterol in A beta-positive nerve terminals from Alzheimers disease cortex. Neurobiol Aging 28: 8 17, 2007. 116. Haeffner F, Smith DG, Barnham KJ, Bush AI. Model studies of cholesterol and ascorbate oxidation by copper complexes: relevance to Alzheimers disease beta-amyloid metallochemistry. J Inorg Biochem 99: 24032422, 2005. 117. Hahnel D, Beyer K, Engelmann B. Inhibition of peroxyl radical-mediated lipid oxidation by plasmalogen phospholipids and alpha-tocopherol. Free Radic Biol Med 27: 10871094, 1999. 118. Hahnel D, Huber T, Kurze V, Beyer K, Engelmann B. Contribution of copper binding to the inhibition of lipid oxidation by plasmalogen phospholipids. Biochem J 340: 377383, 1999. 119. Han X. Lipid alterations in the earliest clinically recognizable stage of Alzheimers disease: implication of the role of lipids in the pathogenesis of Alzheimers disease. Curr Alzheimer Res 2: 65 77, 2005. 120. Han XL, Holtzman DM, McKeel DW. Plasmalogen deciency in early Alzheimers disease subjects and in animal models: molecular characterization using electrospray ionization mass spectrometry. J Neurochem 77: 1168 1180, 2001. 121. Han XL, Holtzman DM, McKeel DW, Kelley J, Morris JC. Substantial sulfatide deciency and ceramide elevation in very early Alzheimers disease: potential role in disease pathogenesis. J Neurochem 82: 809 818, 2002. 122. Hansen LM, Masliah E, Galasko D, Terry RD. Plaque-only Alzheimer-disease is usually the Lewy body variant, and vice-versa. J Neuropath Exp Neurol 52: 648 654, 1993. 123. Hardy J, Allsop D. Amyloid deposition as the central event in the etiology of Alzheimers-disease. Trends Pharmacol Sci 12: 383388, 1991. 124. Hardy JA, Higgins GA. Alzheimers-disease: the amyloid cascade hypothesis. Science 256: 184 185, 1992. 125. Harper JD, Wong SS, Lieber CM, Lansbury PT. Observation of metastable A beta amyloid protobrils by atomic force microscopy. Chem Biol 4: 119 125, 1997. 126. Hashimoto M, Tanabe Y, Fujii Y, Kikuta T, Shibata H, Shido O. Chronic administration of docosahexaenoic acid ameliorates the impairment of spatial cognition learning ability in amyloid betainfused rats. J Nutr 135: 549 555, 2005. 127. Hayashi H, Kimura N, Yamaguchi H, Hasegawa K, Yokoseki T, Shibata M, Yamamoto N, Michikawa M, Yoshikawa Y, Terao K, Matsuzaki K, Lemere CA, Selkoe DJ, Naiki H, Yanagisawa K. A seed for Alzheimer amyloid in the brain. J Neurosci 24: 4894 4902, 2004. 128. Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF, Floyd RA, Buttereld DA. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease. Proc Natl Acad Sci USA 91: 3270 3274, 1994. 129. Hensley K, Hall N, Subramaniam R, Cole P, Harris M, Aksenov M, Aksenova M, Gabbita SP, Wu JF, Carney JM, Lovell MA, Markesbery WR, Buttereld DA. Brain regional correspondence between Alzheimers-disease histopathology and biomarkers of protein oxidation. J Neurochem 65: 2146 2156, 1995. 130. Hertel C, Terzi E, Hauser N, Jakob-Rotne R, Seelig J, Kemp JA. Inhibition of the electrostatic interaction between beta-amyloid peptide and membranes prevents beta-amyloid-induced toxicity. Proc Natl Acad Sci USA 94: 94129416, 1997. 131. Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, 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 Alzheimers disease. J Neurosci 21: 30173023, 2001. 132. Hou LM, Kang I, Marchant RE, Zagorski MG. Methionine 35 oxidation reduces bril assembly of the amyloid A beta-(1-42) peptide of Alzheimers 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 underlies A beta-induced synaptic depression and dendritic spine loss. Neuron 52: 831 843, 2006. 134. Huang XD, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, Scarpa RC, Cuajungco MP, Gray DN, Lim J, Moir RD, Tanzi RE, Bush AI. The A beta peptide of Alzheimers disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry 38: 7609 7616, 1999. 135. Huang XD, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JDA, Hanson GR, Stokes KC, Leopold M, Multhaup G, Goldstein LE, Scarpa RC, Saunders AJ, Lim J, Moir RD, Glabe C, Bowden EF, Masters CL, Fairlie DP, Tanzi RE, Bush AI. Cu(II) potentiation of Alzheimer A beta neurotoxicity: correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem 274: 3711137116, 1999. 136. Hung LW, Ciccotosto GD, Giannakis E, Tew DJ, Perez K, Masters CL, Cappai R, Wade JD, Barnham KJ. Amyloid-beta peptide (A beta) neurotoxicity is modulated by the rate of peptide aggregation: A beta dimers and trimers correlate with neurotoxicity. J Neurosci 28: 11950 11958, 2008. 137. Hutter-Paier B, Huttunen HJ, Puglielli L, Eckman CB, Kim DY, Hofmeister A, Moir RD, Domnitz SB, Frosch MP, Windisch M, Kovacs DM. The ACAT inhibitor CP-113,818 markedly reduces amyloid pathology in a mouse model of Alzheimers disease. Neuron 44: 227238, 2004. 138. Igarashi M, Gao F, Kim HW, Ma KZ, Bell JM, Rapoport SI. Dietary n-6 PUFA deprivation for 15 weeks reduces arachidonic acid concentrations while increasing n-3 PUFA concentrations in organs of post-weaning male rats. BBA Mol Cell Biol Lipids 1791: 132139, 2009. 139. Inoue K, Garner C, Ackermann BL, Oe T, Blair IA. Liquid chromatography/tandem mass spectrometry characterization of oxidized amyloid beta peptides as potential biomarkers of Alzheimers disease. Rapid Commun Mass Spectrom 20: 911 918, 2006. 140. Isaacs AM, Senn DB, Yuan M, Shine JP, Yankner BA. Acceleration of amyloid beta -peptide aggregation by physiological concentrations of calcium. J Biol Chem 281: 27916 27923, 2006. 141. Issa AM, Mojica WA, Morton SC, Traina S, Newberry SJ, Hilton LG, Garland RH, MacLean CH. The efcacy of omega-3 fatty acids on cognitive function in aging and dementia: a systematic review. 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. Truncated -amyloid peptide channels provide an alternative mechanism for Alzheimers Disease and Down syndrome. Proc Natl Acad Sci USA 107: 6838 6543, 2010. 143. Jarrett JT, Berger EP, Lansbury PT. The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimers disease. Biochemistry 32: 4693 4697, 1993. 144. Jeandel C, Nicolas MB, Dubois F, Nabet-Belleville F, Penin F, Cuny G. Lipid peroxidation and free radical scavengers in Alzheimers disease. Gerontology 35: 275282, 1989. 145. Ji SR, Wu Y, Sui SF. Cholesterol is an important factor affecting the membrane insertion of betaamyloid peptide (A beta 1-40), which may potentially inhibit the bril formation. J Biol Chem 277: 6273 6279, 2002. 146. Johansson AS, Garlind A, Berglind-Dehlin F, Karlsson G, Edwards K, Gellerfors P, EkholmPettersson F, Palmblad J, Lannfelt L. Docosahexaenoic acid stabilizes soluble amyloid-beta protobrils and sustains amyloid-beta-induced neurotoxicity in vitro. FEBS J 274: 990 1000, 2007. 147. Kagan BL, Azimov R, Azimova R. Amyloid peptide channels. J Membrane Biol 202: 110, 2004. 148. Kakio A, Nishimoto S, Yanagisawa K, Kozutsumi Y, Matsuzaki K. Cholesterol-dependent formation of GM1 ganglioside-bound amyloid betaprotein, an endogenous seed for Alzheimer amyloid. J Biol Chem 276: 2498524990, 2001. 149. Kakio A, Nishimoto S, Yanagisawa K, Kozutsumi Y, Matsuzaki K. Interactions of amyloid beta-protein with various gangliosides in raft-like membranes: importance of GM1 ganglioside-bound form as an endogenous seed for Alzheimer amyloid. Biochemistry 41: 73857390, 2002. 150. Kakio A, Nishimoto SI, Kozutsumi Y, Matsuzaki K. Formation of a membrane-active form of amyloid beta-protein in raft-like model membranes. Biochem Biophys Res Commun 303: 514 518, 2003.

Downloaded from physiologyonline.physiology.org on January 24, 2012

64

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
151. Kakio A, Yano Y, Takai D, Kuroda Y, Matsumoto O, Kozutsumi Y, Matsuzaki K. Interaction between amyloid beta-protein aggregates and membranes. J Pept Sci 10: 612 621, 2004. 152. Kalvodova L, Kahya N, Schwille P, Ehehalt R, Verkade P, Drechsel D, Simons K. Lipids as modulators of proteolytic activity of BACE: involvement of cholesterol, glycosphingolipids, and anionic phospholipids in vitro. J Biol Chem 280: 3681536823, 2005. 153. Karr JW, Szalai VA. Cu(II) binding to monomeric, oligomeric, and brillar forms of the Alzheimers disease amyloid-beta peptide. Biochemistry 47: 5006 5016, 2008. 154. Kayed R, Sokolov Y, Edmonds B, McIntire TM, Milton SC, Hall JE, Glabe CG. Permeabilization of lipid bilayers is a common conformation-dependent activity of soluble amyloid oligomers in protein misfolding diseases. J Biol Chem 279: 46363 46366, 2004. 155. Kelly BL, Ferreira A. Beta-amyloid-induced dynamin 1 degradation is mediated by N-methyl-Daspartate receptors in hippocampal neurons. J Biol Chem 281: 28079 28089, 2006. 156. Khaselev N, Murphy RC. Susceptibility of plasmenyl glycerophosphoethanolamine lipids containing arachidonate to oxidative degradation. Free Radic Biol Med 26: 275284, 1999. 157. Khaselev N, Murphy RC. Peroxidation of arachidonate containing plasmenyl glycerophosphocholine: facile oxidation of esteried arachidonate at carbon-5. Free Radic Biol Med 29: 620 632, 2000. 158. Khaselev N, Murphy RC. Structural characterization of oxidized phospholipid products derived from arachidonate-containing plasmenyl glycerophosphocholine. J Lipid Res 41: 564 572, 2000. 159. Kim HY. Novel metabolism of docosahexaenoic acid in neural cells. J Biol Chem 282: 18661 18665, 2007. 160. Kitazawa M, Cheng D, LaFerla FM. Chronic copper exposure exacerbates both amyloid and tau pathology and selectively dysregulates cdk5 in a mouse model of AD. J Neurochem 108: 1550 1560, 2009. 161. Komatsu H, Liu L, Murray IV, Axelsen PH. A mechanistic link between oxidative stress and membrane mediated amyloidogenesis revealed by infrared spectroscopy. BBA Biomembranes 1768: 19131922, 2007. 162. Kontush A. Amyloid- : an antioxidant that becomes a pro-oxidant and critically contributes to Alzheimers disease. Free Radic Biol Med 31: 1120 1131, 2001. 163. Kontush A, Berndt C, Weber W, Akopyan V, Arlt S, Schippling S, Beisiegel U. Amyloid- is an antioxidant for lipoproteins in cerebrospinal uid and plasma. Free Radic Biol Med 30: 119 128, 2001. 164. Kontush A, Donarski N, Beisiegel U. Resistance of human cerebrospinal uid to in vitro oxidation is directly related to its amyloid-beta content. Free Radic Res 35: 507517, 2001. 165. Koppaka V, Axelsen PH. Accelerated accumulation of amyloid beta proteins on oxidatively damaged lipid membranes. Biochemistry 39: 10011 10016, 2000. 166. Koppaka V, Paul C, Murray IVJ, Axelsen PH. Early synergy between A 42 and oxidatively damaged membranes in promoting amyloid bril formation by A 40. J Biol Chem 278: 3627736284, 2003. 167. Kremer JJ, Murphy RM. Kinetics of adsorption of beta-amyloid peptide A beta(1-40) to lipid bilayers. J Biochem Biophys Methods 57: 159 169, 2003. 168. Kremer JJ, Pallitto MM, Sklansky DJ, Murphy RM. Correlation of -amyloid aggregate size and hydrophobicity with decreased bilayer uidity of model membranes. Biochemistry 39: 10309 10318, 2000. 169. Kremer JJ, Sklansky DJ, Murphy RM. Prole of changes in lipid bilayer structure caused by betaamyloid peptide. Biochemistry 40: 8563 8571, 2001. 170. Kuczynski B, Reo NV. Evidence that plasmalogen is protective against oxidative stress in the rat brain. Neurochem Res 31: 639 656, 2006. 171. Ladu MJ, Falduto MT, Manelli AM, Reardon CA, Getz GS, Frail DE. Isoform-specic binding of apolipoprotein-E to beta-amyloid. J Biol Chem 269: 2340323406, 1994. 172. LaFerla FM. Calcium dyshomeostasis and intracellular signalling in Alzheimers disease. Nat Rev Neurosci 3: 862 872, 2002. 173. Lal R, Lin H, Quist AP. Amyloid beta ion channel: 3D structure and relevance to amyloid channel paradigm. BBA Biomembranes 1768: 19661975, 2007. 174. Lashuel HA, Hartley D, Petre BM, Walz T, Lansbury PT. Neurodegenerative disease: amyloid pores from pathogenic mutations. Nature 418: 291, 2002. 175. Lashuel HA, Lansbury PT. Are amyloid diseases caused by protein aggregates that mimic bacterial pore-forming toxins? Q Rev Biophys 39: 167 201, 2006. 176. Lau TL, Ambroggio EE, Tew DJ, Cappai R, Masters CL, Fidelio GD, Barnham KJ, Separovic F. Amyloid-beta peptide disruption of lipid membranes and the effect of metal ions. J Mol Biol 356: 759 770, 2006. 177. Lauderback CM, Hackett JM, Huang FF, Keller JN, Szweda LI, Markesbery WR, Buttereld DA. The glial glutamate transporter, GLT-1, is oxidatively modied by 4-hydroxy-2-nonenal in the Alzheimers disease brain: the role of A beta 1 42. J Neurochem 78: 413 416, 2001. 178. Lauderback CM, Kanski J, Hackett JM, Maeda N, Kindy MS, Buttereld DA. Apolipoprotein E modulates Alzheimers A beta(1-42)-induced oxidative damage to synaptosomes in an allele-specic manner. Brain Res 924: 90 97, 2002. 179. Lawson JA, Rokach J, FitzGerald GA. Isoprostanes: formation, analysis and use as indices of lipid peroxidation in vivo. J Biol Chem 274: 2444124444, 1999. 180. Lee SH, Blair IA. Characterization of 4-oxo-2-nonenal as a novel product of lipid peroxidation. Chem Res Tox 13: 698 702, 2000. 181. Lee SJ, Liyanage U, Bickel PE, Xia WM, Lansbury PT, Kosik KS. A detergent-insoluble membrane compartment contains A beta in vivo. Nat Med 4: 730 734, 1998. 182. Lehtonen JYA, Holopainen JM, Kinnunen PKJ. Activation of phospholipase A(2) by amyloid betapeptides in vitro. Biochemistry 35: 94079414, 1996. 183. Leskovjan AC, Lanzirotti A, Miller LM. Amyloid plaques in PSAPP mice bind less metal than plaques in human Alzheimers disease. Neuroimage 47: 12151220, 2009. 184. Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG, Yang A, Gallagher M, Ashe KH. A specic amyloid-beta protein assembly in the brain impairs memory. Nature 440: 352357, 2006. 185. Lim GP, Calon F, Morihara T, Yang FS, Teter B, Ubeda O, Salem N, Frautschy SA, Cole GM. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J Neurosci 25: 30323040, 2005. 186. Lin H, Bhatia R, Lal R. Amyloid beta protein forms ion channels: implications for Alzheimers disease pathophysiology. FASEB J 15: 24332444, 2001. 187. Liu L, Komatsu H, Murray IVJ, Axelsen PH. Promotion of amyloid protein misfolding and brillogenesis by a lipid oxidation product. J Mol Biol 377: 1236 1250, 2008. 188. Liu SJ, Zukin RS. Ca2 -permeable AMPA receptors 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, Sol V, Sommer B, Yankner BA. Amyloid beta interacts with the amyloid precursor protein: a potential toxic mechanism in Alzheimers disease. Nat Neurosci 3: 460 464, 2000. 190. Lovell MA, Ehmann WD, Mattson MP, Markesbery WR. Elevated 4-hydroxynonenal in ventricular uid in Alzheimers disease. Neurobiol Aging 18: 457 461, 1997. 191. Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR. Copper, iron and zinc in Alzheimers disease senile plaques. J Neurol Sci 158: 4752, 1998.

Downloaded from physiologyonline.physiology.org on January 24, 2012

192. Lovell MA, Xie CS, Markesbery WR. Acrolein is increased in Alzheimers disease brain and is toxic to primary hippocampal cultures. Neurobiol Aging 22: 187194, 2001. 193. Lukiw WJ, Cui JG, Marcheselli VL, Bodker M, Botkjaer A, Gotlinger K, Serhan CN, Bazan NG. A role for docosahexaenoic acid-derived neuroprotectin D1 in neural cell survival and Alzheimer disease. J Clin Invest 115: 2774 2783, 2005. 194. Ma JY, Yee A, Brewer HB, Das S, Potter H. Amyloid-associated proteins alpha(1)-antichymotrypsin and apolipoprotein-E promote assembly of Alzheimer beta-protein into laments. Nature 372: 9294, 1994. 195. Maeba R, Sawada Y, Shimasaki H, Takahashi I, Ueta N. Ethanolamine plasmalogens protect cholesterol-rich liposomal membranes from oxidation caused by free radicals. Chem Phys Lipids 120: 145151, 2002. 196. Maeba R, Ueta N. Ethanolamine plasmalogens prevent the oxidation of cholesterol by reducing the oxidizability of cholesterol in phospholipid bilayers. J Lipid Res 44: 164 171, 2003. 197. Mahley RW, Rall SC Jr. Apolipoprotein E: far more than a lipid transport protein. Ann Rev Genom Hum Gen 1: 507537, 2000. 198. Manelli AM, Stine WB, Van Eldik LJ, Ladu MJ. ApoE A 1 42 interactions. J Mol Neurosci 23: 235246, 2004. 199. Marchesi VT. An alternative interpretation of the amyloid A beta hypothesis with regard to the pathogenesis of Alzheimers disease. Proc Natl Acad Sci USA 102: 90939098, 2005. 200. Marcus DL, Thomas C, Rodriguez C, Simberkoff K, Tsai JS, Strafaci JA, Freedman ML. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimers disease. Exp Neurol 150: 40 44, 1998. 201. Markesbery WR. Oxidative stress hypothesis in Alzheimers disease. Free Radic Biol Med 23: 134 147, 1997. 202. Markesbery WR, Carney JM. Oxidative alterations in Alzheimers disease. Brain Pathol 9: 133 146, 1999. 203. Markesbery WR, Lovell MA. Four-hydroxynonenal, a product of lipid peroxidation, is increased in the brain in Alzheimers disease. Neurobiol Aging 19: 3336, 1998. 204. Martinez-Senac MD, Villalain J, Gomez-Fernandez JC. Structure of the Alzheimer beta-amyloid peptide (25-35) and its interaction with negatively charged phospholipid vesicles. Eur J Biochem 265: 744 753, 1999.

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

65

REVIEWS
205. Matsuzaki K. Physicochemical interactions of amyloid -peptide with lipid bilayers. BBA Biomembranes 1768: 19351942, 2007. 206. Matsuzaki K, Horikiri C. Interactions of amyloid beta-peptide (1-40) with ganglioside-containing membranes. Biochemistry 38: 4137 4142, 1999. 207. Matsuzaki K, Noguch T, Wakabayashi M, Ikeda K, Okada T, Ohashi Y, Hoshino M, Naiki H. Inhibitors of amyloid beta-protein aggregation mediated by GM1-containing raft-like membranes. BBA Biomembranes 1768: 122130, 2007. 208. Mattson MP, Cheng B, Culwell AR, Esch FS, Lieberburg I, Rydel RE. Evidence for excitoprotective and intraneuronal calcium-regulating roles for secreted forms of the beta-amyloid precursor protein. Neuron 10: 243254, 1993. 209. Mattson MP, Cheng B, Davis D, Bryant K, Lieberburg I, Rydel RE. -Amyloid peptides destabilize calcium homeostasis and render human cortical neurons vulnerable to excitotoxicity. J Neurosci 12: 376 389, 1992. 210. McGrath LT, McGleenon BM, Brennan S, McColl D, McIlroy S, Passmore AP. Increased oxidative stress in Alzheimers disease as assessed with 4-hydroxynonenal but not malondialdehyde. QJM 94: 485 490, 2001. 211. McIntosh LJ, Trush MA, Troncoso JC. Increased susceptibility of Alzheimers disease temporal cortex to oxygen free radical-mediated processes. Free Radic Biol Med 23: 183190, 1997. 212. McLaurin J, Chakrabartty A. Membrane disruption by Alzheimer beta-amyloid peptides mediated through specic nding to either phospholipids or gangliosides: implications for neurotoxicity. J Biol Chem 271: 2648226489, 1996. 213. McLaurin J, Chakrabartty A. Characterization of the interactions of Alzheimer beta-amyloid peptides with phospholipid membranes. Eur J Biochem 245: 355363, 1997. 214. McLaurin J, Darabie AA, Morrison MR. Cholesterol, a modulator of membrane-associated A beta-brillogenesis. Alz Dis Vasc Etiol Path 977: 376 383, 2002. 215. McLaurin J, Franklin T, Fraser PE, Chakrabartty A. Structural transitions associated with the interaction of Alzheimer beta-amyloid peptides with gangliosides. J Biol Chem 273: 4506 4515, 1998. 216. Minton AP. Implications of macromolecular crowding for protein assembly. Curr Opin Struct Biol 10: 34 39, 2000. 217. Miyata M, Smith JD. Apolipoprotein E allele-specic antioxidant activity and effects on cytotoxicity by oxidative insults and beta-amyloid peptides. Nat Gen 14: 55 61, 1996. 218. Molander-Melin M, Blennow K, Bogdanovic N, Dellheden B, Mansson JE, Fredman P. Structural membrane alterations in Alzheimer brains found to be associated with regional disease development; increased density of gangliosides GM1 and GM2 and loss of cholesterol in detergentresistant membrane domains. J Neurochem 92: 171182, 2005. 219. Monji A, Utsumi H, Ueda T, Imoto T, Yoshida I, Hashioka S, Tashiro K, Tashiro N. The relationship between the aggregational state of the amyloid-beta peptides and free radical generation by the peptides. J Neurochem 77: 14251432, 2001. 220. Montine TJ, Neely MD, Quinn JF, Beal MF, Markesbery WR, Roberts LJ, Morrow JD. Lipid peroxidation in aging brain and Alzheimers disease. 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-4neuroprostanes are not increased in Alzheimers disease. Ann Neurol 52: 175179, 2002. 222. Morandat S, Bortolato M, Anker G, Doutheau A, Lagarde M, Chauvet JP, Roux B. Plasmalogens protect unsaturated lipids against UV-induced oxidation in monolayer. Biochim Biophys Acta 1616: 137146, 2003. 223. Morris MC, Evans DA, Bienias JL, Tangney CC, Bennett DA, Wilson RS, Aggarwal N, Schneider J. Consumption of sh and n-3 fatty acids and risk of incident Alzheimer disease. Arch Neurol 60: 940 946, 2003. 224. Morris MC, Evans DA, Tangney CC, Bienias JL, Wilson RS. Fish consumption and cognitive decline with age in a large community study. Arch Neurol 62: 1849 1853, 2005. 225. Morrow JD, Awad JA, Boss HJ, Blair IA, Roberts LJ, II. Non-cyclooxygenase-derived prostanoids (F2-Isoprostanes) are formed in situ on phosphlipids. Proc Natl Acad Sci USA 89: 1072110725, 1992. 226. Morrow JD, Minton TA, Mukundan CR, Campbell MD, Zackert WE, Daniel VC, Badr KF, Blair IA, Roberts LJ II. Free radical-induced generation of isoprostanes 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, Scheffer PG. Decreased lysophosphatidylcholine/phosphatidylcholine ratio in cerebrospinal uid in Alzheimers disease. J Neural Transm 110: 949 955, 2003. 228. Munishkina LA, Cooper EM, Uversky VN, Fink AL. The effect of macromolecular crowding on protein aggregation and amyloid bril formation. J Mol Recognit 17: 456 464, 2004. 229. Murphy EJ, Huang HM, Cowburn RF, Lannfeltd L, Gibson GE. Phospholipid mass is increased in broblasts bearing the Swedish amyloid precursor mutation. Brain Res Bull 69: 79 85, 2006. 230. Murphy RC. Free-radical-induced oxidation of arachidonoyl plasmalogen phospholipids: antioxidant mechanism and precursor pathway for bioactive eicosanoids. Chem Res Tox 14: 463 472, 2001. 231. Murray IVJ, Liu L, Komatsu H, Uryu K, Xiao G, Lawson JA, Axelsen PH. Membrane mediated amyloidogenesis and the promotion of oxidative lipid damage by amyloid beta proteins. J Biol Chem 282: 93359345, 2007. 232. Murray IVJ, Sindoni ME, Axelsen PH. Promotion of oxidative lipid membrane damage by amyloid beta proteins. Biochemistry 44: 12606 12613, 2005. 233. Namba Y, Tomonaga M, Kawasaki H, Otomo E, Ikeda K. Apolipoprotein-E immunoreactivity in cerebral amyloid deposits and neurobrillary tangles in Alzheimers-disease and Kuru plaque amyloid In Creutzfeldt-Jakob disease. Brain Res 541: 163166, 1991. 234. Natte R, Yamaguchi H, Maat-Schieman MLC, Prins FA, Neeskens P, Roos RAC, van Duinen SG. Ultrastructural evidence of early non-brillar A beta 42 in the capillary basement membrane of patients with hereditary cerebral hemorrhage with amyloidosis, Dutch type. Acta Neuropathol (Berl) 98: 577582, 1999. 235. Nelson TJ, Alkon DL. Oxidation of cholesterol by amyloid precursor protein and beta-amyloid peptide. J Biol Chem 280: 73777387, 2005. 236. Nunomura A, Perry G, Pappolla MA, Friedland RP, Hirai K, Chiba S, Smith MA. Neuronal oxidative stress precedes amyloid-beta deposition in Down syndrome. J Neuropath Exp Neurol 59: 10111017, 2000. 237. Nurk E, Drevon CA, Refsum H, Solvoll K, Vollset SE, Nygard O, Nygaard HA, Engedal K, Tell GS, Smith AD. Cognitive performance among the elderly and dietary sh intake: the Hordaland Health Study. Am J Clin Nutr 86: 1470 1478, 2007. 238. ONuallain B, Shivaprasad S, Kheterpal I, Wetzel R. Thermodynamics of A beta(1-40) amyloid bril elongation. Biochemistry 44: 1270912718, 2005. 239. Okada T, Ikeda K, Wakabayashi M, Ogawa M, Matsuzaki K. Formation of toxic A beta(1-40) brils on GM1 ganglioside-containing membranes mimicking lipid rafts: polymorphisms in A beta(140) brils. J Mol Biol 382: 1066 1074, 2008. 240. Okada T, Wakabayashi M, Ikeda K, Matsuzaki K. Formation of toxic brils of Alzheimers amyloid beta-protein-(1-40) by monosialoganglioside GM1, a neuronal membrane component. J Mol Biol 371: 481 489, 2007. 241. Opazo C, Huang XD, Cherny RA, Moir RD, Roher AE, White AR, Cappai R, Masters CL, Tanzi RE, Inestrosa NC, Bush AI. Metalloenzyme-like activity of Alzheimers disease beta-amyloid- Cu-dependent catalytic conversion of dopamine, cholesterol, and biological reducing agents to neurotoxic H2O2. J Biol Chem 277: 4030240308, 2002. 242. Paltauf F. Ether lipids in biomembranes. Chem Phys Lipids 74: 101139, 1994. 243. Pamplona R, Dalfo E, Ayala VR, Bellmunt MJ, Prat J, Ferrer I, Portero-Otin M. Proteins in human brain cortex are modied by oxidation, glycoxidation, and lipoxidation: effects of Alzheimer disease and identication of lipoxidation targets. J Biol Chem 280: 2152221530, 2005. 244. Patrono C, FitzGerald GA. Isoprostanes: potential markers of oxidant stress in atherothrombotic disease. Arterioscler Thromb Vasc Biol 17: 2309 2315, 1997. 245. Permanne B, Perez C, Soto C, Frangione B, Wisniewski T. Detection of apolipoprotein E dimeric soluble amyloid beta complexes in Alzheimers disease brain supernatants. Biochem Biophys Res Commun 240: 715720, 1997. 246. Petersen CAH, Alikhani N, Behbahani H, Wiehager B, Pavlov PF, Alafuzoff I, Leinonen V, Ito A, Winblad B, Glaser E, Ankarcrona M. The amyloid beta-peptide is imported into mitochondria via the TOM import machinery and localized to mitochondrial cristae. Proc Natl Acad Sci USA 105: 1314513150, 2008. 247. Pettegrew JW, Panchalingam K, Hamilton RL, McClure RJ. Brain membrane phospholipid alterations in Alzheimers disease. Neurochem Res 26: 771782, 2001. 248. Pike CJ, RamezanArab N, Cotman CW. Betaamyloid neurotoxicity in vitro: evidence of oxidative stress but not protection by antioxidants. J Neurochem 69: 16011611, 1997. 249. Pillot T, Goethals M, Vanloo B, Talussot C, Brasseur R, Vandekerckhove J, Rosseneu M, Lins L. Fusogenic properties of the C-terminal domain of the Alzheimer beta-amyloid peptide. J Biol Chem 271: 2875728765, 1996. 250. Plourde M, Fortier M, Vandal M, TremblayMercier J, Freemantle E, Begin M, Pifferi F, Cunnane SC. Unresolved issues in the link between docosahexaenoic acid and Alzheimers disease. Prost Leuk Essen Fatty Acids 77: 301 308, 2007. 251. Pollard HB, Arispe N, Rojas E. Ion channel hypothesis for Alzheimer amyloid peptide neurotoxicity. Cell Molec Neurobiol 15: 513526, 1995. 252. Prasad MR, Lovell MA, Yatin M, Dhillon H, Markesbery WR. Regional membrane phospholipid alterations in Alzheimers disease. Neurochem Res 23: 81 88, 1998. 253. Pratico D, Clark CM, Lee VMY, Trojanowski JQ, Rokach J, FitzGerald GA. Increased 8,12-isoiPF(2 alpha)-VI in Alzheimers disease: correlation of a noninvasive index of lipid peroxidation with disease severity. Ann Neurol 48: 809 812, 2000. 254. Pratico D, Delanty N. Oxidative injury in diseases of the central nervous system: focus on Alzheimers disease. Am J Med 109: 577585, 2000.

Downloaded from physiologyonline.physiology.org on January 24, 2012

66

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
255. Pratico D, Lee VMY, Trojanowski JQ, Rokach J, FitzGerald GA. Increased F-2-isoprostanes in Alzheimers disease: evidence for enhanced lipid peroxidation in vivo. FASEB J 12: 17771783, 1998. 256. Pratico D, Uryu K, Leight S, Trojanowswki JQ, Lee VMY. Increased lipid peroxidation precedes amyloid plaque formation in an animal model of Alzheimer 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. J Clin Invest 115: 2556 2563, 2005. 258. Qahwash IM, Boire A, Lanning J, Krausz T, Pytel P, Meredith SC. Site-specic effects of peptide lipidation on beta-amyloid aggregation and cytotoxicity. J Biol Chem 282: 3698736997, 2007. 259. Quist A, Doudevski L, Lin H, Azimova R, Ng D, Frangione B, Kagan B, Ghiso J, Lal R. Amyloid ion channels: a common structural link for proteinmisfolding disease. Proc Natl Acad Sci USA 102: 1042710432, 2005. 260. Rajendran L, Schneider A, Schlechtingen G, Weidlich S, Ries J, Braxmeier T, Schwille P, Schulz JB, Schroeder C, Simons M, Jennings G, Knolker HJ, Simons K. Efcient inhibition of the Alzheimers disease -secretase by membrane targeting. Science 320: 520 523, 2008. 261. Rangachari V, Moore BD, Reed DK, Sonoda LK, Bridges AW, Conboy E, Hartigan D, Rosenberry TL. Amyloid- (1-42) rapidly forms protobrils and oligomers by distinct pathways in low concentrations of sodium dodecylsulfate. Biochemistry 46: 1245112462, 2007. 262. Reddy PH. Amyloid beta, mitochondrial structural and functional dynamics in Alzheimers disease. Exp Neurol 218: 286 292, 2009. 263. Refolo LM, Pappolla MA, Malester B, LaFrancois J, Bryant-Thomas T, Wang R, Tint GS, Sambamurti K, Duff K. Hypercholesterolemia accelerates the Alzheimers amyloid pathology in a transgenic mouse model. Neurobiol Dis 7: 321331, 2000. 264. Reiss D, Beyer K, Engelmann B. Delayed oxidative degradation of polyunsaturated diacyl phospholipids in the presence of plasmalogen phospholipids in vitro. Biochem J 323: 807 814, 1997. 265. Relini A, Cavalleri O, Rolandi R, Gliozzi A. The two-fold aspect of the interplay of amyloidogenic proteins with lipid membranes. Chem Phys Lipids 158: 19, 2009. 266. Roberts LJ, Fessel JP. The biochemistry of the isoprostane, neuroprostane, and isofuran pathways of lipid peroxidation. Chem Phys Lipids 128: 173186, 2004. 267. Roberts LJ, Montine TJ, Markesbery WR, Tapper AR, Hardy P, Chemtob S, Dettbarn WD, Morrow JD. Formation of isoprostane-like compounds (neuroprostanes) in vivo from docosahexaenoic acid. J Biol Chem 273: 1360513612, 1998. 268. Roberts LJ, II, Morrow JD. IsoProstanes: novel markers of endogenous lipid peroxidation and potential mediators of oxidant injury. Ann NY Acad Sci 744: 237242, 1994. 269. Roheim PS, Carey M, Forte T, Vega GL. Apolipoproteins in human cerebrospinal uid. Proc Natl Acad Sci USA 76: 4646 4649, 1979. 270. Rottkamp CA, Nunomura A, Raina AK, Sayre LM, Perry G, Smith MA. Oxidative stress, antioxidants, and Alzheimer disease. Alzheimer Dis Assoc Disord 14, Suppl 1: S62S66, 2000. 271. Sanan DA, Weisgraber KH, Russell SJ, Mahley RW, Huang D, Saunders A, Schmechel D, Wisniewski T, Frangione B, Roses AD, Strittmatter WJ. Apolipoprotein-E associates with beta-amyloid peptide of Alzheimers-disease to form novel monobrils: isoform Apoe4 associates more Efciently than Apoe3. J Clin Invest 94: 860 869, 1994. 272. Sarell CJ, Syme CD, Rigby SEJ, Viles JH. Copper(II) binding to amyloid-beta brils of Alzheimers disease reveals a picomolar afnity: stoichiometry and coordination geometry are independent of A beta oligomeric form. Biochemistry 48: 4388 4402, 2009. 273. Sayre LM, Perry G, Harris PLR, Liu YH, Schubert KA, Smith MA. In situ oxidative catalysis by neurobrillary tangles and senile plaques in Alzheimers disease: a central role for bound transition metals. J Neurochem 74: 270 279, 2000. 274. Sayre LM, Zelasko DA, Harris PLR, Perry G, Salomon RG, Smith MA. 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimers disease. J Neurochem 68: 20922097, 1997. 275. Schaefer EJ, Bongard V, Beiser AS, Lamon-Fava S, Robins SJ, Au R, Tucker KL, Kyle DJ, Wilson PWF, Wolf PA. Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham heart study. Arch Neurol 63: 15451550, 2006. 276. Schauerte JA, Wong PT, Wisser KC, Ding H, Steel DG, Gafni A. Simultaneous single molecule uorescence and conductivity studies reveal distinct classes of A species on lipid bilayers. Biochemistry 49: 30313039, 2010. 277. Schippling S, Kontush A, Arlt S, Buhmann C, Sturenburg HJ, Mann U, Muller-Thomsen T, Beisiegel U. Increased lipoprotein oxidation in Alzheimers disease. Free Radic Biol Med 28: 351360, 2000. 278. Schmechel DE, Saunders AM, Strittmatter WJ, Crain BJ, Huletter CM, Joo SH, Pericakvance MA, Goldgaber D, Roses AD. Increased amyloid betapeptide deposition in cerebral-cortex as a consequence of apolipoprotein-E genotype in lateonset Alzheimer-disease. Proc Natl Acad Sci USA 90: 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 loss exceeds amyloid plaque load in a transgenic mouse model of Alzheimers disease. Am J Pathol 164: 14951502, 2004. 280. Schneeweis LA, Koppaka V, Lund-Katz S, Phillips MC, Axelsen PH. Structural analysis of lipoprotein E particles. Biochemistry 44: 1252512534, 2005. 281. Sengupta P, Garai K, Sahoo B, Shi Y, Callaway DJE, Maiti S. The amyloid beta peptide [A beta(140)] is thermodynamically soluble at physiological concentrations. 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 protein dimers isolated directly from Alzheimers brains impair synaptic plasticity and memory. Nat Med 14: 837 842, 2008. 283. Shao H, Jao S, Ma K, Zagorski MG. Solution structures of micelle-bound amyloid beta-(1-40) and beta-(1-42) peptides of Alzheimers disease. J Mol Biol 285: 755773, 1999. 284. Shearer J, Szalai VA. The Amyloid- peptide of Alzheimers disease binds Cu in a linear Bis-His coordination environment: insight into a possible neuroprotective mechanism for the amyloidpeptide. J Am Chem Soc 130: 17826 17835, 2008. 285. Shin SJ, Lee SE, Boo JH, Kim M, Yoon YD, Kim SI, Mook-Jung I. Proling proteins related to amyloid deposited brain of Tg2576 mice. Proteomics 4: 3359 3368, 2004. 286. Shringarpure R, Grune T, Sitte N, Davies KJA. 4-Hydroxynonenal-modied amyloid-beta peptide inhibits the proteasome: possible importance in Alzheimers disease. Cell Mol Life Sci 57: 18021809, 2000. 287. Siegel SJ, Bieschke J, Powers ET, Kelly JW. The oxidative stress metabolite 4-hydroxynonenal promotes Alzheimer protobril formation. Biochemistry 46: 15031510, 2007. 288. Simons M, Keller P, De Strooper B, Beyreuther K, Dotti CG, Simons K. Cholesterol depletion inhibits the generation of beta-amyloid in hippocampal neurons. Proc Natl Acad Sci USA 95: 6460 6464, 1998. 289. Sjogren M, Blennow K. The link between cholesterol and Alzheimers disease. World J Biol Psych 6: 8597, 2005. 290. Smith DP, Ciccotosto GD, Tew DJ, Fodero-Tavoletti MT, Johanssen T, Masters CL, Barnham KJ, Cappai R. Concentration dependent Cu2 induced aggregation and dityrosine formation of the Alzheimers disease amyloid- peptide. Biochemistry 46: 28812891, 2007. 291. Smith DP, Smith DG, Curtain CC, Boas JF, Pilbrow JR, Ciccotosto GD, Lau TL, Tew DJ, Perez K, Wade JD, Bush AI, Drew SC, Separovic F, Masters CL, Cappai R, Barnham KJ. Copper mediated amyloid-beta toxicity is associated with an intermolecular histidine bridge. J Biol Chem 281: 1514515154, 2006. 292. Smith MA, Casadesus G, Joseph JA, Perry G. Amyloid-beta and tau serve antioxidant functions in the aging and Alzheimer brain. Free Radic Biol Med 33: 1194 1199, 2002. 293. Smith MA, Harris PLR, Sayre LM, Beckman JS, Perry G. Widespread peroxynitrite-mediated damage in Alzheimers disease. J Neurosci 17: 26532657, 1997. 294. Smith MA, Hirai K, Hsiao K, Pappolla MA, Harris PLR, Siedlak SL, Tabaton M, Perry G. Amyloidbeta deposition in Alzheimer transgenic mice is associated with oxidative stress. J Neurochem 70: 22122215, 1998. 295. Smith MA, Perry G, Richey PL, Sayre LM, Anderson VE, Beal MF, Kowall N. Oxidative damage in Alzheimers. Nature 382: 120 121, 1996. 296. Smith MA, Taneda S, Richey PL, Miyata S, Yan SD, Stern D, Sayre LM, Monnier VM, Perry G. Advanced Maillard reaction end-products are associated with Alzheimer-disease pathology. Proc Natl Acad Sci USA 91: 5710 5714, 1994. 297. Sokolov Y, Kozak JA, Kayed R, Chanturiya A, Glabe C, Hall JE. Soluble amyloid oligomers increase bilayer conductance by altering dielectric structure. J Gen Physiol 128: 637 647, 2006. 298. Song WL, Lawson JA, Reilly D, Rokach J, Chang CT, Giasson B, FitzGerald GA. Neurofurans, novel indices of oxidant stress derived from docosahexaenoic 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 Alzheimers disease-associated amyloid -protein is an antimicrobial peptide. PLoS ONE 5: e9505, 2010. 300. Sparks DL, Scheff SW, Hunsaker JC, Liu HC, Landers T, Gross DR. Induction of Alzheimer-like beta-amyloid immunoreactivity in the brains of rabbits with dietary-cholesterol. Exp Neurol 126: 88 94, 1994.

Downloaded from physiologyonline.physiology.org on January 24, 2012

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

67

REVIEWS
301. Stewart CR, Wilson LM, Zhang Q, Pham CLL, Waddington LJ, Staples MK, Stapleton D, Kelly JW, Howlett GJ. Oxidized cholesterol metabolites found in human atherosclerotic lesions promote apolipoprotein C-II amyloid bril formation. Biochemistry 46: 55525561, 2007. 302. Stokin GB, Lillo C, Falzone TL, Brusch RG, Rockenstein E, Mount SL, Raman R, Davies P, Masliah E, Williams DS, Goldstein LSB. Axonopathy and transport decits early in the pathogenesis of Alzheimers disease. Science 307: 12821288, 2005. 303. Stratman NC, Castle CK, Taylor BM, Epps DE, Melchior GW, Carter DB. Isoform-specic interactions of human apolipoprotein E to an intermediate conformation of human Alzheimer amyloidbeta peptide. Chem Phys Lipids 137: 52 61, 2005. 304. Strausak D, Mercer JFB, Dieter HH, Stremmel W, Multhaup G. Copper in disorders with neurological symptoms: Alzheimers, Menkes, and Wilson diseases. Brain Res Bull 55: 175185, 2001. 305. Streltsov VA. X-ray absorption and diffraction studies of the metal binding sites in amyloid beta-peptide. Eur Biophys J 37: 257263, 2008. 306. Streltsov VA, Titmuss SJ, Epa VC, Barnham KJ, Masters CL, Varghese JN. The structure of the amyloid-beta peptide high-afnity copper II binding site in Alzheimer disease. Biophys J 95: 34473456, 2008. 307. Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS, Pericakvance M, Schmechel D, Saunders AM, Goldgaber D, Roses AD. Binding of human apolipoprotein-E to synthetic amyloid-beta peptide: isoform-specic effects and implications for late-onset Alzheimerdisease. Proc Natl Acad Sci USA 90: 8098 8102, 1993. 308. Subasinghe S, Unabia S, Barrow CJ, Mok SS, Aguilar MI, Small DH. Cholesterol is necessary both for the toxic effect of A beta peptides on vascular smooth muscle cells and for A beta binding to vascular smooth muscle cell membranes. J Neurochem 84: 471 479, 2003. 309. Sultana R, Buttereld DA. Oxidatively modied GST and MRP1 in Alzheimers disease brain: implications for accumulation of reactive lipid peroxidation products. Neurochem Res 29: 2215 2220, 2004. 310. Surewicz WK, Mantsch HH. The conformation of dynorphin A-(1-13) in aqueous solution as studied by Fourier transform infrared spectroscopy. J Mol Struct 214, 143147, 1989. 311. Svennerholm L, Gottfries CG. Membrane-lipids, selectively diminished in Alzheimer brains, suggest synapse loss as a primary event in early-onset form (type-I) and demyelination in late-onset form (typeII). J Neurochem 62: 1039 1047, 1994. 312. Tamaoka A, Miyatake F, Matsuno S, Ishii K, Nagase S, Sahara N, Ono S, Mori H, Wakabayashi K, Tsuji S, Takahashi H, Shoji S. Apolipoprotein E allele-dependent antioxidant activity in brains with Alzheimers disease. Neurol 54: 2319 2321, 2000. 313. Terzi E, Holzemann G, Seelig J. Alzheimer betaamyloid peptide 25-35: electrostatic interactions with phospholipid membranes. Biochemistry 33: 7434 7441, 1994. 314. Terzi E, Holzemann G, Seelig J. Self-association of beta-amyloid peptide (1-40) in solution and binding to lipid membranes. J Mol Biol 252: 633 642, 1995. 315. Terzi E, Holzemann G, Seelig J. Interaction of Alzheimer beta-amyloid peptide(1-40) with lipid membranes. Biochemistry 36: 1484514852, 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 the Alzheimer disease amyloid- peptide results in abolition of superoxide dismutase-like structures and inhibition of neurotoxicity. J Biol Chem 280: 1335513363, 2005. 317. Tien M, Berlett BS, Levine RL, Chock PB, Stadtman ER. Peroxynitrite-mediated modication of proteins at physiological carbon dioxide concentration: 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, Takuma H, Ohnishi K, Ishibashi K, Teraoka R, Sakama N, Yamashita T, Nishitsuji K, Ito K, Shimada H, Lambert MP, Klein WL, Mori H. A mouse model of amyloid oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. J Neurosci 30: 4845 4856, 2010. 319. Torp R, Head E, Milgram NW, Hahn F, Ottersen OP, Cotman CW. Ultrastructural evidence of brillar beta-amyloid associated with neuronal membranes in behaviorally characterized aged dog brains. Neuroscience 96: 495506, 2000. 320. Townsend M, Shankar GM, Mehta T, Walsh DM, Selkoe DJ. Effects of secreted oligomers of amyloid beta-protein on hippocampal synaptic plasticity: a potent role for trimers. J Physiol 572: 477 492, 2006. 321. Treiber C, Quadir MA, Voigt P, Radowski M, Xu SJ, Munter LM, Bayer TA, Schaefer M, Haag R, Multhaup G. Cellular copper import by nanocarrier systems, intracellular availability, and effects on amyloid beta peptide secretion. Biochemistry 48: 4273 4284, 2009. 322. Trojanowski JQ, Lee VM. The role of tau in Alzheimers 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 serum cholesteryl ester-docosahexaenoic acid levels in Alzheimers disease: a case-control study. Br J Nutr 89: 483 489, 2003. 324. Uchida K. Current status of acrolein as a lipid peroxidation product. Trends Cardiovasc Med 9: 109 113, 1999. 325. Uchida K. 4-Hydroxy-2-nonenal: a product and mediator 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 lipid peroxidation reaction: formation of free acrolein and its conjugate with lysine residues in oxidized low 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, Noguchi N, Niki E, Osawa T. Protein-bound acrolein: potential markers for oxidative stress. Proc Natl Acad Sci USA 95: 4882 4887, 1998. 328. Uchida K, Stadtman ER. Modication of histidineresidues in proteins by reaction with 4-hydroxynonenal. Proc Natl Acad Sci USA 89: 4544 4548, 1992. 329. Usui K, Hulleman JD, Paulsson JF, Siegel SJ, Powers ET, Kelly JW. Site-specic modication of Alzheimers peptides by cholesterol oxidation products enhances aggregation energetics and neurotoxicity. Proc Natl Acad Sci USA 106: 1856318568, 2009. 330. Van Nostrand WE, Melchor JP, Rufni L. Pathologic amyloid beta-protein cell surface bril assembly on cultured human cerebrovascular smooth muscle cells. J Neurochem 70: 216 223, 1998. 331. Varadarajan S, Yatin S, Aksenova M, Buttereld DA. Alzheimers amyloid beta-peptide-associated free radical oxidative stress and neurotoxicity. J Struct Biol 130: 184 208, 2000. 332. Varadarajan S, Yatin S, Buttereld DA. Alzheimers amyloid beta peptide (1-42) brils are not always neurotoxic. Alzheimers Rep 3: 7176, 2000. 333. Wahrle SE, Jiang H, Parsadanian M, Hartman RE, Bales KR, Paul SM, Holtzman DM. Deletion of Abca1 increases A beta deposition in the PDAPP transgenic mouse model of Alzheimer disease. J Biol Chem 280: 43236 43242, 2005. 334. Wakabayashi M, Matsuzaki K. Formation of amyloids by a beta-(1-42) on NGF-differentiated PC12 cells: roles of gangliosides and cholesterol. J Mol Biol 371: 924 933, 2007. 335. Wakabayashi M, Okada T, Kozutsumi Y, Matsuzaki K. GM1 ganglioside-mediated accumulation of 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. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature 416: 535539, 2002. 337. Walsh DM, Lomakin A, Benedek GB, Condron MM, Teplow DB. Amyloid beta-protein brillogenesis: detection of a protobrillar intermediate. J Biol Chem 272: 22364 22372, 1997. 338. Wang SSS, Rymer DL, Good TA. Reduction in cholesterol and sialic acid content protects cells from 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 denes Abeta-lipid interactions. J Biol Chem 276: 3356133568, 2001. 340. Weisgraber KH, Shinto LH. Identication of the disulde-linked homodimer of apolipoprotein E3 in plasma: impact on receptor-binding activity. J Biol Chem 266: 12029 12034, 1991. 341. Widenbrant MJO, Rajadas J, Sutardja C, Fuller GG. Lipid-induced -amyloid peptide assemblage fragmentation. Biophys J 91: 4071 4080, 2006. 342. Williams TI, Lynn BC, Markesbery WR, Lovell MA. Increased levels of 4-hydroxynonenal and acrolein, neurotoxic markers of lipid peroxidation, in the brain in mild cognitive impairment and early Alzheimers disease. Neurobiol Aging 27: 1094 1099, 2006. 343. Wisniewski T, Castano EM, Golabek A, Vogel T, Frangione B. Acceleration of Alzheimers bril formation by apolipoprotein-E in-vitro. Am J Pathol 145: 1030 1035, 1994. 344. Wong PT, Schauerte JA, Wisser KC, Ding H, Lee EL, Steel DG, Gafni A. Amyloid-beta membrane binding and permeabilization are distinct processes inuenced separately by membrane charge and uidity. J Mol Biol 386: 8196, 2009. 345. Wu Y, Luo Y. Transgenic C. elegans as a model in Alzheimers research. Curr Alzheimer Res 2: 37 47, 2005. 346. Yamaguchi H, Maat-Schieman MLC, van Duinen SG, Prins FA, Neeskens P, Natte R, Roos RAC. Amyloid beta protein (A beta) starts to deposit as plasma membrane-bound form in diffuse plaques of brains from hereditary cerebral hemorrhage with amyloidosis-Dutch type, Alzheimer disease and nondemented aged subjects. J Neuropath Exp Neurol 59: 723732, 2000. 347. Yamazaki T, Chang TY, Haass C, Ihara Y. Accumulation and aggregation of amyloid beta-protein in late endosomes of Niemann-Pick type C cells. J Biol Chem 276: 4454 4460, 2001.

Downloaded from physiologyonline.physiology.org on January 24, 2012

68

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

REVIEWS
348. Yanagisawa K, Odaka A, Suzuki N, Ihara Y. GM1 ganglioside-bound amyloid beta-protein (A beta): a possible form of preamyloid in Alzheimers disease. Nat Med 1: 10621066, 1995. 349. Yang DS, Smith JD, Zhou ZM, Gandy SE, Martins RN. Characterization of the binding of amyloidbeta peptide to cell culture-derived native apolipoprotein E2, E3, and E4 isoforms and to isoforms from human plasma. J Neurochem 68: 721725, 1997. 350. Yao JK, Wengenack TM, Curran GL, Poduslo JF. Reduced membrane lipids in the cortex of Alzheimers disease transgenic mice. Neurochem Res 34: 102108, 2009. 351. Yao ZX, Drieu K, Szweda LI, Papadopoulos V. Free radicals and lipid peroxidation do not mediate beta-amyloid-induced neuronal cell death. Brain Res 847: 203210, 1999. 352. Yip CM, Darabie AA, McLaurin J. A beta 42peptide assembly on lipid bilayers. J Mol Biol 318: 97107, 2002. 353. Yip CM, McLaurin J. Amyloid-beta peptide assembly: a critical step in brillogenesis and membrane disruption. Biophys J 80: 1359 1371, 2001. 354. Yoshiike Y, Kayed R, Milton SC, Takashima A, Glabe CG. Pore-forming proteins share structural and 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 disrupt amyloid beta-aggregation and their effects on amyloid-beta cytotoxicity. J Biol Chem 276: 3229332299, 2001. 356. Yoshimoto N, Tasaki M, Shimanouchi T, Umakoshi H, Kuboi R. Oxidation of cholesterol catalyzed by amyloid beta-peptide (A beta)-Cu complex on lipid membrane. J Biosci Bioeng 100: 455 459, 2005. 357. Zagol-Ikapitte I, Masterson TS, Amarnath V, Montine TJ, Andreasson KI, Boutaud O, Oates JA. Prostaglandin H-2-derived adducts of proteins correlate with Alzheimers disease severity. J Neurochem 94: 1140 1145, 2005. 358. Zaman Z, Roche S, Fielden P, Frost PG, Niriella DC, Cayley AC. Plasma concentrations of vitamins A and E and carotenoids in Alzheimers disease. Age Ageing 21: 9194, 1992. 359. Zerbinatti CV, Wahrle SE, Kim H, Cam JA, Bales K, Paul SM, Holtzman DM, Bu GJ. Apolipoprotein E and low density lipoprotein receptor-related protein facilitate intraneuronal A beta 42 accumulation in amyloid model mice. J Biol Chem 281: 36180 36186, 2006. 360. Zhang QH, Powers ET, Nieva J, Huff ME, Dendle MA, Bieschke J, Glabe CG, Eschenmoser A, Wentworth P, Lerner RA, Kelly JW. Metaboliteinitiated protein misfolding may trigger Alzheimers disease. Proc Natl Acad Sci USA 101: 4752 4757, 2004. 361. Zhou S, Zhou H, Walian PJ, Jap BK. Regulation of -secretase activity in Alzheimers disease. Biochemistry 46: 25532563, 2007. 362. Zommara M, Tachibana N, Mitsui K, Nakatani N, Sakono M, Ikeda I, Imaizumi K. Inhibitory effect of ethanolamine plasmalogen on iron-dependent and copper-dependent lipid-peroxidation. Free Radic Biol Med 18: 599 602, 1995. 363. Zou K, Gong JS, Yanagisawa K, Michikawa M. A novel function of monomeric amyloid beta-protein serving as an antioxidant molecule against metal-induced oxidative damage. J Neurosci 22: 4833 4841, 2002. 364. Zubenko GS. Hippocampal membrane-alteration in Alzheimers-disease. Brain Res 385: 115121, 1986.

Downloaded from physiologyonline.physiology.org on January 24, 2012

PHYSIOLOGY Volume 26 February 2011 www.physiologyonline.org

69

Вам также может понравиться