review article the impact of cholesterol, dha, and ...family mainly involved in the cholesterol...

17
Hindawi Publishing Corporation BioMed Research International Volume 2013, Article ID 814390, 16 pages http://dx.doi.org/10.1155/2013/814390 Review Article The Impact of Cholesterol, DHA, and Sphingolipids on Alzheimerā€™s Disease Marcus O. W. Grimm, 1,2,3 Valerie C. Zimmer, 1 Johannes Lehmann, 1 Heike S. Grimm, 1 and Tobias Hartmann 1,2,3 1 Experimental Neurology, Saarland University, Kirrberger Street 1, 66421 Homburgr, Saar, Germany 2 Neurodegeneration and Neurobiology, Saarland University, Kirrberger Street 1, 66421 Homburg, Germany 3 Deutsches Institut fĀØ ur DemenzPrĀØ avention (DIDP), Saarland University, Kirrberger Street 1, 66421 Homburgr, Saar, Germany Correspondence should be addressed to Marcus O. W. Grimm; [email protected] and Tobias Hartmann; [email protected] Received 30 April 2013; Accepted 13 July 2013 Academic Editor: Cheng-Xin Gong Copyright Ā© 2013 Marcus O. W. Grimm et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Alzheimerā€™s disease (AD) is a devastating neurodegenerative disorder currently aļ¬€ecting over 35 million people worldwide. Pathological hallmarks of AD are massive amyloidosis, extracellular senile plaques, and intracellular neuroļ¬brillary tangles accompanied by an excessive loss of synapses. Major constituents of senile plaques are 40ā€“42 amino acid long peptides termed -amyloid (A). A is produced by sequential proteolytic processing of the amyloid precursor protein (APP). APP processing and A production have been one of the central scopes in AD research in the past. In the last years, lipids and lipid-related issues are more frequently discussed to contribute to the AD pathogenesis. is review summarizes lipid alterations found in AD postmortem brains, AD transgenic mouse models, and the current understanding of how lipids inļ¬‚uence the molecular mechanisms leading to AD and A generation, focusing especially on cholesterol, docosahexaenoic acid (DHA), and sphingolipids/glycosphingolipids. 1. APP Processing Amyloid plaques are composed of aggregated amyloid- pep- tides, derived from sequential proteolytic processing of the amyloid-precursor protein (APP), a type-I transmembrane protein with a large extracellular N-terminal domain and a short intracellular C-terminal tail [1]. APP and its gene family members, the APP-like proteins APLP1 and APLP2, are highly conserved proteins expressed in numerous species and tissues pointing out their physiological importance [2]. Indeed, triple knockout of APP/APLP1/APLP2 in mice results in postnatal lethality involving brain development abnormalities and cortical dysplasia [3]. In contrast, sin- gle knockout of APP has a minor phenotype consisting of reduced body weight [2], commissure defects [4], and hypersensitivity to epileptic seizures [5] demonstrating the mutual functional compensation of the gene family members and additionally illustrates the physiological role of APP. Furthermore, a potential contribution to the formation of dendritic and synaptic structures as well as in long-term po- tentiation (LTP) [6ā€“8] suggests a possible impact on cognitive function. APP can be cleaved in two distinct pathways, the amyl- oidogenic and nonamyloidogenic pathways. e non-amyloi- dogenic processing of APP by -secretases avoids the forma- tion of A peptides by cleaving inside the A domain [9]. ereby, the large N-terminal ectodomain -secreted APP (sAPP) is released into the extracellular matrix, whereas the short C-terminal part (-CTF) remains within the membrane for further processing. Notably, sAPP has neuroprotective and memory enhancing properties [10, 11], hypothesizing that sAPP might mediate a major physiological function of APP [12]. e -secretases belonging to the ADAM family (a disintegrin and metalloprotease), in particular ADAM10 and ADAM17 (tumor necrosis factor converting enzyme/TACE), have emerged as predominant -secretases [13ā€“15]. Like APP itself, these proteins are type I integral membrane proteins.

Upload: others

Post on 30-Jan-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Hindawi Publishing CorporationBioMed Research InternationalVolume 2013, Article ID 814390, 16 pageshttp://dx.doi.org/10.1155/2013/814390

    Review ArticleThe Impact of Cholesterol, DHA, and Sphingolipids onAlzheimerā€™s Disease

    Marcus O. W. Grimm,1,2,3 Valerie C. Zimmer,1 Johannes Lehmann,1

    Heike S. Grimm,1 and Tobias Hartmann1,2,3

    1 Experimental Neurology, Saarland University, Kirrberger Street 1, 66421 Homburgr, Saar, Germany2Neurodegeneration and Neurobiology, Saarland University, Kirrberger Street 1, 66421 Homburg, Germany3Deutsches Institut fuĢˆr DemenzPraĢˆvention (DIDP), Saarland University, Kirrberger Street 1, 66421 Homburgr, Saar, Germany

    Correspondence should be addressed to Marcus O. W. Grimm; [email protected] andTobias Hartmann; [email protected]

    Received 30 April 2013; Accepted 13 July 2013

    Academic Editor: Cheng-Xin Gong

    Copyright Ā© 2013 Marcus O. W. Grimm et al. This is an open access article distributed under the Creative Commons AttributionLicense, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properlycited.

    Alzheimerā€™s disease (AD) is a devastating neurodegenerative disorder currently affecting over 35 million people worldwide.Pathological hallmarks of AD are massive amyloidosis, extracellular senile plaques, and intracellular neurofibrillary tanglesaccompanied by an excessive loss of synapses. Major constituents of senile plaques are 40ā€“42 amino acid long peptides termedš›½-amyloid (Aš›½). Aš›½ is produced by sequential proteolytic processing of the amyloid precursor protein (APP). APP processing andAš›½ production have been one of the central scopes in AD research in the past. In the last years, lipids and lipid-related issues aremore frequently discussed to contribute to the AD pathogenesis.This review summarizes lipid alterations found in AD postmortembrains, AD transgenic mouse models, and the current understanding of how lipids influence the molecular mechanisms leading toAD and Aš›½ generation, focusing especially on cholesterol, docosahexaenoic acid (DHA), and sphingolipids/glycosphingolipids.

    1. APP Processing

    Amyloid plaques are composed of aggregated amyloid-š›½ pep-tides, derived from sequential proteolytic processing of theamyloid-precursor protein (APP), a type-I transmembraneprotein with a large extracellular N-terminal domain anda short intracellular C-terminal tail [1]. APP and its genefamily members, the APP-like proteins APLP1 and APLP2,are highly conserved proteins expressed in numerous speciesand tissues pointing out their physiological importance[2]. Indeed, triple knockout of APP/APLP1/APLP2 in miceresults in postnatal lethality involving brain developmentabnormalities and cortical dysplasia [3]. In contrast, sin-gle knockout of APP has a minor phenotype consistingof reduced body weight [2], commissure defects [4], andhypersensitivity to epileptic seizures [5] demonstrating themutual functional compensation of the gene family membersand additionally illustrates the physiological role of APP.Furthermore, a potential contribution to the formation of

    dendritic and synaptic structures as well as in long-term po-tentiation (LTP) [6ā€“8] suggests a possible impact on cognitivefunction.

    APP can be cleaved in two distinct pathways, the amyl-oidogenic andnonamyloidogenic pathways.Thenon-amyloi-dogenic processing of APP by š›¼-secretases avoids the forma-tion of Aš›½ peptides by cleaving inside the Aš›½ domain [9].Thereby, the large N-terminal ectodomain š›¼-secreted APP(sAPPš›¼) is released into the extracellular matrix, whereas theshort C-terminal part (š›¼-CTF) remainswithin themembranefor further processing. Notably, sAPPš›¼ has neuroprotectiveand memory enhancing properties [10, 11], hypothesizingthat sAPPš›¼ might mediate a major physiological functionof APP [12]. The š›¼-secretases belonging to the ADAMfamily (a disintegrin and metalloprotease), in particularADAM10 and ADAM17 (tumor necrosis factor š›¼ convertingenzyme/TACE), have emerged as predominant š›¼-secretases[13ā€“15]. Like APP itself, these proteins are type I integralmembrane proteins.

  • 2 BioMed Research International

    The amyloidogenic pathway, on the other hand, is ini-tiated by the š›½-secretase BACE1 (beta-site APP cleavingenzyme) that generates soluble š›½-secreted APP (sAPPš›½)and the membrane-tethered fragment š›½-CTF. BACE1 is amembrane-bound aspartyl protease belonging to the pepsinfamily, expressed in neurons [16]. The main š›½-secretaseactivity is found in the secretory pathway including the Golgicompartments, secretory vesicles, and endosomes [17, 18].Apart from its amyloidogenic effect, less is known about thephysiological function of BACE1; however, in BACE1 knock-out mice, myelination is affected [19, 20]. Initial cleavageof APP by š›¼-secretase in the non-amyloidogenic pathway,or by š›½-secretase in the amyloidogenic pathway, is typicallyfollowed by š›¾-secretase processing, a multimeric complexconsisting of at least four subunitsā€”presenilin 1 (PS1) orpresenilin 2 (PS2), anterior pharynx defective 1 homologue(APH1), presenilin enhancer 2 (PEN2), and nicastrin [21]with PS1/PS2 being the catalytic centre [22, 23]. Importantly,mutations inside the PS genes are responsible for early onsetAD [24]. PS1 and PS2 are multitransmembrane spanningaspartyl proteases cleaving the C-terminal stubs š›¼- andš›½-CTF within the centre of their transmembrane domain[25], generating p3 and Aš›½ peptides of varying length (e.g.,Aš›½38, Aš›½40, and Aš›½42). Among the Aš›½ species generated,hydrophobic Aš›½42 peptides self-aggregate to small oligomersbefore being manifested as senile plaques composed of adense core of amyloid fibrils [26, 27]. Simultaneously, theAPP intracellular domain (AICD), which is discussed toregulate gene transcription, is released into the cytosol [28].The substrate APP and the secretases involved in APPcleavage are all transmembrane proteins, suggesting that thesurrounding lipid microenvironment may play a pivotal rolein the pathogenesis of the disease.

    2. Cholesterol and AD

    The human brain has a very high cholesterol content, mainlyassociated with myelin. Due to the limited transport overthe blood-brain barrier (BBB), the brain cholesterol levelis largely independent of the serum cholesterol concentra-tions. The vast majority of brain cholesterol is providedby glial de novo synthesis. Noteworthy, cholesterol trans-port between neurons and glia cells is mostly providedby clusterin/apolipoprotein J (ApoJ) and apolipoprotein E(ApoE) containing lipoproteins. Intriguingly, the ApoEšœ€4allele genotype is the predominant genetic risk factor forAD, whereas the šœ€2 allele seems to be protective and šœ€3being the most common form. A possible explanation forthis might be reduced Aš›½-clearance and/or increased for-mation of amyloid in the presence of the šœ€4 genotype,but many other mechanisms have also been suggested [29ā€“31]. Recent genome-wide association studies (GWASs) havegreatly extended our knowledge on AD risk genes. Interest-ingly, two main functional risk clusters were identified. MostGWAS-identified risk genes are either linked to inflammationor to lipid/membrane processes. Besides ApoE polymor-phism, single-nucleotide polymorphisms for clusterin (CLU),ABCA7, and PICALMwere discovered to raise the individualrisk for developingAD [32ā€“34].On one hand, clusterin serves

    as a major lipid binding protein [35]. Interestingly, clustrinmRNA and protein levels are increased in AD, thereby, cor-relating with disease severity [36, 37]. Furthermore, clusterinmay be involved in modulation of apoptosis, inflammation,and Aš›½ aggregation [38ā€“40]. ABCA7, belonging to the ATP-binding cassette transporters, is expressed throughout thebrain and especially in hippocampal neurons [41, 42]. ABCA7is involved in cholesterol efflux from cells to ApoE and affectsAPP processing [43]. These genetic links between AD riskand cholesterol are supported by strong epidemiological evi-dence linking hypercholesterolemia with dementia [44, 45].Likemany other life-style influencedAD risk factors, elevatedblood cholesterol levels, even if only moderately increased,are most relevant if present at midlife [46]. Additionally,elevated 24OH-cholesterol levels were observed in the serumof AD patients [47].

    Cholesterol is an essential component of mammalian cellmembranes. Based on its extraordinary structure, consistingof a fused rigid ring system, a polar hydroxyl group, and ahydrocarbon tail, cholesterol is essential for bilayerā€™s functionand organisation. Due to the impact of the rigid ring system,cholesterol can increase the order within the membraneand thereby affects membrane fluidity. Especially in lipidmicrodomains, envisioned as so-called ā€œlipid rafts,ā€ primarilyfound in the plasma membrane, the trans-Golgi, and endo-somal membranes, this feature is extremely important. Lipidrafts are strongly enriched in sphingomyelin, glycosphin-golipids, and cholesterol. Cholesterol provides tight packingof the lipids in these microdomains. Some membrane pro-teins are preferentially sited in these ordered microdomains.Mechanistically important is the transient colocalization ofAPP with the amyloidogenic secretases, š›½-secretase, and š›¾-secretase, in the lipid rafts, pointing out that within theamyloidogenic pathway the close colocalization of theseproteins is, at least partly, mediated by cholesterol [48, 49].Nonamyloidogenic processing and theš›¼-secretases, however,are localized outside the lipid rafts [50]. This characteristicimplies the possible regulation of the nonamyloidogenic andamyloidogenic pathways by altered membrane cholesterolamounts. Indeed, evidence suggests that cholesterol tends toform complexes with š›½-CTF and potentially with full-lengthAPP.Thereby, the translocation ofš›½-CTF/APP is promoted tothe cholesterol-enriched microdomains and thus providingthem to the amyloidogenic processing [51, 52]. Additionally,it is well established that cholesterol directly stimulatesš›½- andš›¾-secretases; vice versa cholesterol depletion by cyclodextrinor statins leads to a decreased activity [53ā€“55]. Recently, itwas described that statins influence APP maturation andphosphorylation not by cholesterol lowering but by theloss of cholesterol precursors [56]. Remarkably, cholesterolfurther affects the thickness of the lipid bilayer which directlyinfluences the š›¾-secretase cleavage activity and specificity.With increasing membrane thickness, smaller Aš›½ species(e.g., Aš›½38 and Aš›½40) were preferentially produced, whereasAš›½42/43 only occurred in smaller amounts [57]. Addition-ally, š›¼-secretase cleavage is increased by lowering cholesterollevels. This effect was attributed to increased membrane flu-idity and impaired APP internalisation. After treatment with

  • BioMed Research International 3

    lovastatin, ADAM10 protein level was elevated [50]. Asidefrom its impact on the Aš›½ generation, cholesterol might evenmodify Aš›½ aggregation and the subsequent neurotoxicity.Under physiological conditions, mainly monomeric Aš›½ pep-tides develop fromAPP processing [58].Though, in AD thesemonomers are prone to form oligomers, protofibrils, andfibrils, whereby the oligomers are considered to be the mosttoxic subspecies [59]. It was highlighted that cholesterol-enriched microdomains promote amyloid aggregation, whiledepletion of cholesterol leads to reduced aggregation [60].Recent findings implicated that cholesterol facilitates š›½-sheetformation by direct interaction with Phe19 amino acid of theAš›½ peptide [61]. In line with these findings, some studiesshowed a relationship between elevated cholesterol level andincreased toxicity of Aš›½ oligomers [62, 63]. Contradictory tothis, other investigations reported cholesterol to be protective[64, 65]. Hence, although cholesterol is essential for humanbrain development and function, it can as well be attributedto a potent role in mediating aggregation and neurotoxicity.

    In line with the strong impact of cholesterol on APPprocessing, there is evidence that the cleavage products ofAPP themselves affect cholesterol metabolism. In mouseembryonic fibroblasts deficient inAPP/APLP2āˆ’/āˆ’ or PS1/2āˆ’/āˆ’and therefore unable to produce Aš›½, cholesterol levels werehighly elevated. Interestingly, these cholesterol aberrationswere abrogated by treatment with Aš›½1-40 displaying anegative feed-back cycle of Aš›½1-40 by inhibiting the 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR), therate-limiting step in cholesterol de novo synthesis [66, 67].Additionally, decreased membrane fluidity and increasedcholesterol content in lipid rafts were observed in cellsdevoid of Aš›½ [68]. Within the APP family of proteins, APPapparently has an especially important role for cholesterolhomeostasis. In APP knock-out mice, cholesterol is stronglyincreased [66, 69], and APP knockout has a number of addi-tional cholesterol- and lipid-related consequences, includingreduced diet-dependent atherosclerosis [70] and increasedNiemann-Pick cholesterol phenotype [71]. To some part, thisappears to be caused by a lack of AICD, resulting in alteredLRP1 levels [72]. LRP1 belongs to a lipoprotein-receptorfamily mainly involved in the cholesterol uptake of neu-rons, whereby cholesterol is provided by ApoE-containinglipoproteins [72]. Changes in membrane cholesterol contentwill trigger homeostatic actions affecting the regulation andmembrane content of many other lipids. Accordingly, manyother lipids affect the cholesterol regulation and several ofthese lipids are targeted by APP/Aš›½ or themselves change Aš›½production.

    Taking all of these findings into consideration (Figure 1;See Table S1 in Supplementary Material available onlineat http://dx.doi.org/10.1155/2013/814390), pharmacologicalintervention in cholesterol metabolism might be a possibletarget in AD treatment. It seems likely that cholesterollowering by administration of HMGCR inhibitors (e.g.,statins) might refer to reduced Aš›½ levels accompanied byslower cognitive decline and improved mental status.

    Animal model studies associated hypercholesteremiawith elevated Aš›½ levels [80ā€“82]. On the opposite, reduced Aš›½

    accumulation was achieved after administration of choles-terol lowering drugs (e.g., statins), underlining their potentialrole in the disease treatment [83ā€“86]. However, one studyreported unaffected Aš›½ levels, whereas another one evenobserved increased Aš›½ deposition [87, 88]. Interestingly, thelatter alteration was only detected in female mice. Thesedeviations might be contributed to differences in animalmodels, used drugs, and period of drug administration. Apotential beneficial effect of cholesterol lowering drugs wasfurther investigated in observational studies and randomizedcontrolled trials, whereas many observational studies withhigh number of participants associated statin intake withreduced development of AD [89ā€“91], others exhibited nodifferences [92, 93]. However, recent randomized, double-blind, placebo-controlled studies reported no benefit of statinuse in individuals suffering from mild to moderate AD [94,95]. Since in these trials patients were already affected byAD, statins might be ascribed to a more preventive thantherapeutic function. Thus, further studies have to evaluatea protective effect in earlier clinical stages.

    3. Docosahexaenoic Acid andAlzheimerā€™s Disease

    Docosahexaenoic acid (DHA) is an essential šœ”-3 polyun-saturated fatty acid (PUFA) mainly found in marine food,especially in oily fish. Only a small amount of DHA canbe produced endogenously by synthesis out of š›¼-linoleicacid through elongation and desaturation [96], whereas themain DHA is provided by dietary intake. Approximately60% of the unsaturated fatty acids in neuronal membranesconsist of DHA, thus, representing the most common šœ”-3 fatty acid in the human brain. DHA rapidly incorporatesinto phospholipids of cellular membranes and changes themembrane fluidity by the formation of highly disordereddomains concentrated in PUFA-containing phospholipidsbut depleted in cholesterol (reviewed in [97]). Especiallyin synapses, these alterations in membrane fluidity play animportant role in neurotransmission, ion channel formation,and synaptic plasticity. This suggests a potential role of DHAin memory, learning, and cognitive processes. In young rats,the administration of DHA leads to an improved learningability [98]. Additionally, DHA is involved in neuronal dif-ferentiation [99], neurogenesis [100], and protection againstsynaptic loss [101].Therefore,DHA is discussed to be involvedin pathological processes of AD.

    DHA is decreased in certain regions of AD postmortembrains, like pons, white matter and, in particular, frontal greymatter, and hippocampus [102]. Additionally, peroxidationproducts of DHA, which is very susceptible to lipid peroxi-dation due to its six double bounds, are elevated in AD brainshypothesizing that DHA loss can be ascribed to the increasedoxidative stress [103]. These lipid alterations seem to occurnot only in advanced AD patients but also in early stages ofthe disease [104].

    Several epidemiological studies tried to correlate DHAplasma levels or the amount of dietary intaken fish withcognitive decline. The Rotterdam study presented the first

  • 4 BioMed Research International

    Statins

    N-terminus

    C-terminus

    APP

    Aš›½

    Amyloidogenicprocessing

    š›½-secretase š›¾-secretase

    Aš›½

    HOCholesterol

    HMG-CoA reductase

    Shift towards lipid rafts ā†‘

    -BACE1 protein level ā†‘-Secretase activity ā†‘

    -PS1/PS2 gene expressionā†‘-Secretase activity ā†‘

    Aggregation ā†‘

    Cholesterol de novo synthesis

    š›¼-secretase

    Nonamyloidogenicprocessing

    -ADAM10 gene expressionā†“

    -sAPPš›¼ level/secretase activity ā†“

    Aš›½

    Toxic oligomers ā†‘

    Figure 1: Schematic representation of the proposed mechanisms of cholesterol on APP processing and Aš›½ aggregation.

    findings about an inverse correlation between increased fishintake and all-cause dementia [105]. However, reexaminationwith a longer follow-up period found no association betweendietary intake of šœ”-3 fatty acids and dementia [106]. Never-theless, further studies like the PAQUID study [107] or theCHAP study [108] supported the protective effect of fish orDHA consumption initially found in the Rotterdam study.These effects were even more pronounced in individuals notcarrying the ApoE šœ€4 allele [109, 110].

    Beside the epidemiological studies, findings from animalmodels and in vitro experiments suggest that increaseddietary intake of DHA is associated with a reduced riskof AD. In a 3xTg-AD mouse model, that exhibits bothAš›½ and tau pathologies [111], DHA supplementation causeda significant reduction in soluble and intraneuronal Aš›½levels as well as tau phosphorylation [112]. Similar resultswere obtained with APPSwedish transgenic mice, revealingsignificant plaque reduction in the hippocampus and parietalcortex accompanied by alterations of APP cleavage products[113]. In AD-model rats, produced by infusion of Aš›½1-40 intothe cerebral ventricle, DHA also improved learning ability[114]. In vitro, Aš›½ fibrillation was found to be decreased inthe presence of DHA [73].

    Recently, we and others elucidated the molecular mech-anisms involved in the reduced Aš›½ burden in the presenceof DHA (summarized in Table 1). In the neuroblastoma cellline SH-SY5Y, DHA directly inhibited amyloidogenic š›½- andš›¾-secretase activities, resulting in a dose-dependent reduc-tion of Aš›½ levels. On the other hand, non-amyloidogenicAPP processing was increased in DHA-treated cells, as weobserved elevated sAPPš›¼ levels caused by an enhancedADAM17 protein stability [74]. Interestingly, DHA exhibited

    various interactions with cholesterol homeostasis. Beside adirect inhibition of the HMGCR, DHA induced a cholesterolshift from lipid raft to the nonraft fractions, illustrating analternative secretase activity modulating pathway [74]. Inline with these findings the DHA-mediator NPD1, derivedfrom DHA processing by cytosolic phospholipase A2 and 15-lipooxygenase, is known to directly affect APP processingresulting in elevated sAPPš›¼ and lower sAPPš›½ and accord-ingly Aš›½ levels [75, 76]. Interestingly, phospholipase A2 and15-lipooxygenase and as a consequence NPD1 were foundto be reduced in the hippocampus of AD patients and inAD mouse models [75, 76]. Beside the observed shift ofAPP processing from the amyloidogenic pathway to the non-amyloidogenic pathway, NPD1 also acts neuroprotective bydownregulating inflammatory signalling, apoptosis, and Aš›½-induced neurotoxicity [76, 115].

    These findings indicate a possible therapeutic use ofDHA in preventing, modulating, or improving AD progres-sion. Clinical trials, however, delivered ambiguous resultsconcerning DHA supplementation and cognitive functionssuggesting a limited benefit depending on the disease stageand ApoE allele genotype [116ā€“118]. Further investigationswith great cohorts have to clarify whether DHA has morepreventing than therapeutic effects by including only patientsin the earliest stages of memory decline. More recently, anadvanced DHA formulation containing several diet-derivedmolecules to enhance DHA activity resulted in a number ofclinical trials that resulted in various degrees of cognitivebenefit. The benefit was most pronounced in patients withvery mild to mild AD but less in mild to moderate ADpatients [119, 120]. Most recently, at the ADPD conference2012, Scheltens reported on an open-label extension study in

  • BioMed Research International 5

    Table 1: Summary of mechanisms of DHA and DHA derivateson APP processing. DHA both affects amyloidogenic and non-amyloidogenic pathways via multiple mechanisms resulting in adecrease in Aš›½ production. In opposite to cholesterol it has beenreported that DHA decreases Aš›½ aggregation and toxicity. Directeffects of DHA on APP processing are further enhanced by a DHA-mediated decrease in cholesterol de novo synthesis [73ā€“79].

    (a) Effect of DHA

    Affected pathway Mechanism of actionNonamyloidogenicprocessing

    sAPPš›¼ ā†‘ADAM 17 protein stability ā†‘

    Amyloidogenic processing

    Aš›½ ā†“š›½-Secretase activity ā†“Endosomal BACE1 ā†“š›¾-Secretase activity ā†“PS1 shift: raft ā†’ non-raft

    Aš›½ Oligomerization andtoxicity

    Aš›½ Fibrillation ā†“Soluble toxic oligomers ā†“Aš›½ Phagocytosis ā†‘

    Cholesterol homeostasisHMG-CoA reductase activity ā†“Cholesterol de novo synthesis ā†“Cholesterol shift: raft ā†’ non-raft

    Other non-APP-mediatedpathways/mechanisms

    SorLA/R11 ā†‘, a sorting proteinreduced in ADNeuronal differentiation ā†‘Protection against synaptic loss,Synaptogenesis ā†‘Neurogenesis ā†‘Inflammation ā†“Reactive oxidative species ā†“

    (b) Effect of DHA derivates

    Affected pathway Mechanism of action

    NPD1

    Nonamyloidogenicprocessing

    sAPPš›¼ ā†‘ADAM 10 maturation ā†‘

    Amyloidogenicprocessing

    Aš›½ ā†“sAPPš›½ ā†“BACE 1 protein level ā†“

    Aš›½ Toxicity Neuroprotective and antiapoptoticSoluble toxic oligomers ā†“

    very mild to mild AD patients with continuous increase inmemory performance over the study period of 48weeks [121].

    4. Sphingo- and Glycosphingolipids

    Sphingolipids are a heterogeneous group of lipids, struc-turally based on the 18-carbon amino alcohol sphingosinewhich is synthesized from palmitoyl-CoA and serine byserine palmitoyl-CoA transferase (SPT), representing therate-limiting step in sphingolipid synthesis and shown to beregulated by AICD [122]. Ceramide (Cer) is an importantbranching point for the synthesis of different sphingolipid

    subspecies. For example, sphingomyelin (SM) is generatedout of Cer by SM-synthase, whereas the neutral sphin-gomyelinase (nSMase) catalyzes the turnover of SM to Cer.Glycosylation of Cer results in the production of glycosphin-golipids which can be further processed to gangliosides. Thecerebroside synthase adds a galactose moiety to Cer whichis the first step towards the formation of sulfatides. Finally,the decomposition of Cer is provided by the ceramidasereceiving sphingosine which itself can be phosphorylated tosphingosine-1-phosphate (S1P). First evidence of sphingolipidmetabolism being involved in neurodegenerative diseaseswas derived from investigations of lysosomal storage diseases(LSDs). This group of inherited metabolic disorders is char-acterized by accumulation of different sphingolipids due todysfunction or deficiency of the corresponding lysosomalenzymes. Importantly, affected patients clinically developprogressive cognitive decline resulting in early dementia.Additionally, AD-related pathologies like Aš›½ accumulationand hyperphosphorylation of tau, leading to neurofibrillarytangles, can be observed [123, 124]. Beside these similaritiesbetween AD and LSD, several studies of AD postmortembrains indicate that the sphingolipid metabolism is alteredduring AD progression, further substantiated by biochemicalstudies, linking sphingolipids to APP processing [125ā€“131](Figure 2).

    4.1. Ceramide. The majority of postmortem brain tissueanalysis found elevated Cer levels in the grey and whitematters of AD patients. These alterations were observedeven in early stages of AD hypothesizing that these mightpromote the development of the disease [125, 128, 131, 132].In line with these findings, gene expression abnormalities ofthe key enzymes that control sphingolipid metabolism werefound in AD patients: enzymes involved in glycosphingolipidsynthesis (e.g., UDP-glucose ceramide glucosyltransferase)were altered accompanied by changes in enzymes resultingin the accumulation of Cer (e.g., serine palmitoyltransferase,neutral sphingomyelinase, and acid sphingomyelinase) [131,133]. Recently, Mielke et al. reported in a 9-year-follow-up study that even elevated baseline serum Cer levels areassociated with a higher risk (up to 10-fold) of developing AD[134], indicating that serum Cer is associated with incidentAD.

    In contrast to the results obtained from AD postmortembrains, the analysis of AD mouse models revealed incon-sistent data. Cer levels are elevated in the cerebral cortexof APPSL/PS1Ki transgenic mice, whereas the correspondingsingle-transgenic mice did not show this alteration [135].Furthermore, in APPSL/PS1M146L transgenic mice, in whichthe time course of pathology is closer to that seen inmost cur-rently available models, Cer did not accumulate in disease-associated brain regions (cortex and hippocampus) [136].These different findings might be attributed to the variousmouse models used in these studies [135, 136]. The authorssuggest that APPSL/PS1Kimice, compared to the othermousemodels, produce exceeding amounts of N-truncated Aš›½x-42,also found in AD brains [135].

    It is well established that ceramides induce apoptosisand exhibit neurotoxic properties [137ā€“139]. Interestingly,

  • 6 BioMed Research International

    Nonamyloidogen

    ic pathway

    Amyloidogenic pathway

    GM1

    p3 Aš›½

    Aš›½

    š›¼-CTF

    š›¼-cleavage

    š›¾-cleavage

    APP

    AICD

    APP

    mat

    urat

    ion

    Gangliosides

    Prot

    ein

    Ceramides

    SPT

    AIC

    D

    š›¾-s

    ecre

    tase

    Aš›½š›½-CTF

    Aš›½ degradation and clear

    ance

    Toxic oligomersGM1

    nSM

    ase

    Ceramides

    Cell death

    Sphingosin-1-P

    Aš›½

    Aš›½

    Aš›½ Aš›½

    Oligomerizatio

    n

    Sphingomyelin

    Via ApoESulfatides

    stabi

    lity

    š›½-s

    ecre

    tase

    sAPP

    š›¼

    sAPP

    š›½

    Figure 2: Schematic illustration of the effects of sphingolipids and glycosphingolipids on APP processing. Interestingly, APP processingin return affects the metabolic pathways of sphingolipids. For example, it has been shown that AICD regulates the sphingolipid de novosynthesis by decreasing the expression of the Serinepalmitoyl-CoA-Transferase (SPT) or that Aš›½ itself directly increases the activity of thesphingomyelin degrading enzyme Sphingomyelinase (SMase), resulting in complex regulatory cycles which are dysregulated in the case ofAlzheimerā€™s disease.

    Aš›½ toxicity is linked to Cer-dependent apoptotic pathways.Lee et al. observed an Aš›½-induced elevation of nSMase andconsequently increased Cer levels which resulted in remark-able cell death. Inhibiting this pathway abolished the Aš›½-triggered cascade [140]. In this context, activation of nSMaseby Aš›½42 but not Aš›½40 has been reported [66, 141] (Figure 2).Recently, a potential novel mechanism of ceramide-enrichedexosomes released by Aš›½-treated astrocytes was proposed tobe responsible for Aš›½-induced apoptosis. Thereby, nSMase2was essential for charging these exosomes with Cer [142,143]. Beside the involvement of Cer in Aš›½ toxicity, Cer hasbeen shown to alter APP processing and Aš›½ production.Increasing Cer levels by either direct Cer administration orstimulation of endogenous biosynthesis by nSMase resultedin enhanced Aš›½ production. Elevated Aš›½ levels are attributedto Cer-induced protein stabilization of š›½-secretase BACE1,whereas š›¾-secretase is not affected [144]. Further studieselucidated the underlying mechanism of increased BACE1stability: elevated Cer level caused upregulation of the acetyl-transferases, ATase1, and ATase2, acetylating BACE1 proteinand thus protecting the nascent protein from degradation[145]. Taken together, it seems likely that Cer is the drivingforce in a circulus vitiosus: increasing Cer levels lead to anintensified Aš›½ production whereupon Aš›½ is responsible forCer accumulation.

    Noteworthy in this context, S1P is discussed as a possiblecounterpart. S1P is considered to be neuroprotective andimportant for neuronal differentiation [146, 147]. Remark-ably, one study reported reduced S1P levels in frontotemporal

    grey matter of AD patients [131], which implicates a possiblerole in AD. However, recent findings reported an increasedproteolytic activity of BACE1 by direct interaction withS1P [148]. Therefore, additional studies addressing S1P areimportant to clarify the significance of S1P in APP processingand AD.

    4.2. Sphingomyelin. Sphingomyelin is an important compo-nent of mammalian cell membranes, particularly enrichedin myelin sheets and especially represented in the brain[149]. Considering the observations outlined in the preced-ing chapter it is suggested that SM concentrations in ADbrains might be decreased. Analysis of postmortem brain,however, show inconsistent results. Although two studiesreported elevated SM levels in AD brains compared to age-related controls [150, 151], two studies described a signif-icant decrease [131, 132]. Further studies only revealed amodest reduction in severe AD postmortem brains and nosignificant differences in earlier stages of the disease [125].Moreover, recent investigations found increased SM levels inthe cerebrospinal fluid (CSF) of individuals suffering fromprodromal AD, whereas there was only a slight but notsignificant decrease in mild and moderate AD groups [152].Interestingly, a current epidemiological study correlatedhigher plasma SM levels with slower cognitive decline amongAD patients, illustrating SM as potential sensitive blood-based biomarker for disease progression [153]. Importantly,nSMase was reported to be upregulated in AD brains [133],

  • BioMed Research International 7

    resulting in increased SM breakdown. Additionally, in cellculture studies, nSMase activity is known to be elevated inpresenilin familial Alzheimerā€™s disease mutations (PS-FAD)causing early onset AD [66], pointing towards a possiblerole of SMases in sporadic late onset as well as in familialearly onset AD pathology. Interestingly, SM itself is discussedto alter APP processing. Increasing SM by either directtreatment of cells with SM or inhibition of nSMase resultedin diminished Aš›½ levels [66]. In this context, it is again worthmentioning that Aš›½42 itself regulates SM homeostasis asdescribed previously.

    4.3. Gangliosides. Gangliosides are a family of sialic acidcontaining glycosphingolipids, highly expressed in neuronaland glial membranes, where they play important roles fordevelopment, proliferation, differentiation, and maintenanceof neuronal tissues and cells [154, 155]. The first step towardsthe formation of gangliosides is the glycosylation of Cerby glucosylceramide-synthase (GCS). According to theirnumber of sialic acid residues, gangliosides are separated infour different ganglio-series: o-series, a-series, b-series, andc-series. Importantly, the most common brain gangliosidesbelong either to the a-series (GM1 and GD1a) or b-series(GD1b and GT1b).The ganglioside GM3 serves as a commonprecursor for a- and b-series gangliosides.TheGD3-synthase(GD3S) catalyzes the synthesis of GD3 by adding sialic acid toGM3, segregating the a- and b-series of gangliosides [156] andtherefore controlling the levels of the major brain ganglio-sides. Gangliosides are also able to interact with furthermem-brane lipids like SM and cholesterol, thereby, being involvedin the formation of lipid rafts [157]. Interestingly, postmortemstudies of AD brains suggest a strong connection betweenganglioside homeostasis and AD pathology. In a previouswork, Kracun et al. found all major brain gangliosides to bereduced in the temporal and frontal cortex and in nucleusbasalis of Meynert, whereas simple gangliosides GM2 andGM3 were elevated in parietal and frontal cortex [158]. GM3elevation was further supported by a recent study. Here, theauthors also described an increase in glucosylceramide levels,the precursor for ganglioside synthesis [159]. Additionally,Gottfries et al. reported a significant reduction of gangliosidesin the grey matter of early onset AD subjects comparedto late onset AD and control individuals. Nevertheless, adecrease in total gangliosides was also observed in brainsof late onset AD patients, however, to a smaller extent[160]. A more recent study found elevated levels of GM1and GM2 in lipid raft fractions of the temporal and frontalcortex of AD brains [161]. Moreover, the analysis of ADtransgenic mouse models suggests an altered gangliosidemetabolism in AD. Barrier et al. compared different trans-genic mice with age-matched wildtype controls. While allmice expressing APP (SL) showed an increase in GM2 andGM3 in the cerebral cortex and a moderate decrease incomplex b-series gangliosides, only APP/PS1Ki transgenicmice exhibited a loss of complex a-series gangliosides, GT1a,GD1a, and GM1 [162]. In summary, ganglioside metabolismseems to be highly affected during disease progression.Whilemore complex gangliosides appear to be depleted, simplegangliosides, like GM1 andGM3 are increased. Asmentioned

    previously GM3 is an important precursor for a- and b-seriesgangliosides suggesting a disease-dependent alteration in thebiosynthesis of these ganglioside series. Indeed, we found aclose link between APP processing products and gangliosidemetabolism.The activity of GD3S, the key enzyme convertinga- to b-series gangliosides, is significantly reduced by twoseparate and additive mechanisms. On one hand, GD3Sactivity is inhibited by the binding of Aš›½ peptides to GM3,consequently reducing substrate availability and preventingthe conversion of GM3 to GD3. On the other hand, genetranscription of GD3S is downregulated and mediated by theAPP intracellular domain (AICD), thus, resulting in GD3depletion and GM3 accumulation [163].

    Furthermore, especially GM1 is related to several AD-specific pathomechanisms like altered APP processing,aggregation, and cytotoxicity. GM1 was found to decreasesAPPš›¼ levels and to increase Aš›½ generation, whereassAPPš›½ levels were unchanged [164]. This suggests increasedš›¾-secretase and decreased š›¼-secretase activities withoutaffecting š›½-secretase. Indeed, as recently described, thedirect administration of gangliosides to purified š›¾-secretaseresulted in an increased enzyme activity. Moreover, a shifttowards the formation of Aš›½42 peptides was observed [165].Interestingly, Aš›½ decreases membrane fluidity by bindingGM1. As a consequence, amyloidogenic APP processingwas stimulated, proposing an Aš›½-triggered, GM1-mediated,vicious cycle [166]. Further mechanisms linking ganglio-sides to Aš›½ production were described by Tamboli et al.Inhibition of GCS, the committed step towards gangliosideformation, significantly decreased Aš›½ formation. An alteredAPP maturation and cell surface transport, leading to lessaccess of APP to amyloidogenic processing in the endosomalcompartments, were proposed as the underlyingmechanisms[167].

    Furthermore, GM1 seems to be particularly importantas a ā€œseedā€ for amyloid plaque formation. Thereby, GM1interacts with Aš›½, resulting in GAš›½ complexes [168]. Thiscomplex tends to aggregate more easily due to changing thesecondary structure of Aš›½ towards š›½-sheet formation [169,170]. Interestingly, Mahfoud et al. described a sphingolipidbinding domain of Aš›½, which is also contained in HIV-1 and prion proteins [171]. Further investigations displayedaccumulation and aggregation of Aš›½ on cell membranesespecially in GM1-enriched lipid rafts, resulting in enhancedcytotoxicity [172, 173]. Additionally, enhanced cytotoxicity ofAš›½ fibrils was observed after release of GM1 from damagedneurons, indicating a possible aggravation mechanism [174].Importantly, GAš›½ complexes also occur in AD brains andaged mice [175]. Synaptosomes, prepared from aged mousebrains, exhibited GM1 clusters, showing high ability toinitiate Aš›½ aggregation [176]. Remarkably, Aš›½ depositionwas observed to begin mainly at the presynaptic neuronalmembranes in AD brains, suggesting a possible role of GAš›½in the early pathogenesis of AD [177, 178].

    4.4. Sulfatides. Another sphingolipid subgroup, possiblyinvolved in AD pathogenesis, are sulfatides. They are gener-ated from Cer by adding a galactose moiety, catalyzed by theceramide galactosyltransferase (CGT). Finally, synthesis of

  • 8 BioMed Research International

    sulfatides is provided by cerebroside sulfotransferase (CST),which transfers a sulfate group to the galactosyl moiety. Thedegradation of sulfatides takes place in the lysosomal com-partment, where arylsulfatase A (ASA) hydrolyzes the sulfategroup. ASA deficiency leads to accumulation of sulfatidesand to the clinical picture of metachromatic leukodystrophy.Sulfatides are especially enriched in myelin sheaths makingup about 5% of the myelin lipids. Hence, they are particularlyproduced by oligodendrocytes and Schwann cells. Never-theless, sulfatides have also been detected in neurons andastrocytes, however, in a lower amount [179]. Interestingly,AD pathology is known to induce focal demyelination anddegeneration of oligodendrocytes [180].

    Importantly, Han et al. described an extraordinary deple-tion of sulfatide levels in all analyzed brain regions of ADsubjects. Sulfatides were depleted up to 93% in grey matterand up to 58% in white matter.These alterations were alreadyobserved in the earliest stages of the disease [125]. Addition-ally, a previous study found decreased sulfatide levels in thewhite matter of the frontal lobe only in patients with lateonset AD compared to age-matched controls and early onsetADpatients [160]. Furthermore, Bandaru et al. also describeddecreased sulfatide content in the white matter; however,they did not confirm the alterations in the grey matter [150].In contrast, further studies found no significant changesbetween control andADbrains [132, 159].Worthmentioning,a 40% reduction in CSF sulfatide levels was detected amongAD patients. In this context, sulfatide/phosphatidylinositolratio was proposed as potential clinical biomarker for earlyAD diagnosis [181]. However, data obtained by analyzingtransgenic mouse models in respect to alterations in sulfatidelevels are more inconsistent. Although Barrier et al. foundno significant changes between wildtype mice and doubletransgenic APPSL/PS1Ki mice or single-transgenic PS1Kimice [135], a more recent study found significantly decreasedsulfatide levels in the forebrain of these mouse models[159].

    Noteworthy, there is a possible link between sulfatidehomeostasis and ApoE trafficking. First, the intercellular sul-fatide transport in the brain is mediated by ApoE-containinglipoproteins. Interestingly, humanApoE4 carrying transgenicmice presented the highest sulfatide depletion in the brain incomparison to wildtype ApoE or human ApoE3 transgenicmice [181]. Furthermore, a recent study showed a significant,age-related decrease of sulfatides in APP transgenic mice,which was completely abolished by ApoE knockout [182].These findings offer a possible explanation for decreased sul-fatide levels in AD brains. On the other hand, sulfatides seemto be involved in the ApoE-dependent Aš›½ clearance. Directsupplementation of sulfatides to cultured cells dramaticallyreduced Aš›½ levels. This observation was most likely ascribedto a modification of Aš›½ clearance through an endocytoticpathway. Thereby, sulfatides facilitated the ApoE-mediatedAš›½ clearance, especially of ApoE4 containing vesicles [183].Nevertheless, the importance of sulfatide-dependent molec-ular mechanisms, being involved in AD pathogenesis, is stillambiguous.Therefore, further investigations are necessary toclarify the role of sulfatides in APP processing and AD.

    5. Lipids and Tau Pathology

    Beside amyloid plaques, neurofibrillary tangles (NFTs), con-sisting of hyperphosphorylated tau proteins, are consideredto be a pivotal pathological hallmark of AD [184ā€“186].Although this review focuses on the impact of lipids on APPprocessing, it is worth mentioning that tau pathology is alsoaffected by an altered lipid homeostasis. Tau proteins belongto the family of microtubule-associated proteins, importantfor the assembly of tubulin monomers into microtubules, tostabilize the neuronal microtubule network which is essentialfor maintaining cell shape and axonal transport [187]. InAD, this function is disrupted, due to the hyperphospho-rylation of serine/threonine residues of tau. This abnormalphosphorylation promotes the release of tau proteins frommicrotubules, its disassembly, and self-assembly into pairedhelical filaments (PHFs), a major component of NFTs, and,as a consequence, provokes microtubule disruption [188ā€“191]. Supporting the connection between lipids and tau,Kawarabayashi et al. reported an accumulation of phosphory-lated tau in brain-extracted lipid rafts of an ADmouse model[192]. In addition, tau phosphorylation by cyclin-dependentkinase 5 (CDK5) was observed in lipid rafts after short-term incubation of SH-SY5Y cells with Aš›½ peptides [193].Interestingly, Niemann-Pick type C (NPC), an inheritedlysosomal storage disease with an abnormal intracellularaccumulation of cholesterol, also exhibits tau pathology [194,195]. Noteworthy, these alterations are more pronounced inneurons containing higher cholesterol levels [194, 196]. Inline with these findings elevated Aš›½ and total tau levelswere observed in the cerebrospinal fluid of NPC patients[197]. In an NPC mouse model, Sawamura et al. illustrateda possible explanation for these hyperphosphorylated tauforms. They found an intensified activation of the mitogen-activated protein kinase (MAPK)-pathway, one of severalkinases phosphorylating tau physiologically in the brain[198]. Not only in NPCmodel mice but also in further mousemodels, the cholesterol and ApoE status were associated withincreased tau phosphorylation [199ā€“201]. In contrast, statintreatment reduced NFT burden in normocholesterolemicand hypercholesterolemic mice. This effect, however, wasrather attributed to the anti-inflammatory properties thanto the cholesterol lowering aspect of statins [202]. Addition-ally, simvastatin treatment of hypercholesterolemic subjectswithout dementia revealed a significant phospho-tau-181decrease in the CSF, whereas no differences in total tauor Aš›½ levels were observed [203]. Importantly, membranecholesterol levels are closely linked to theAš›½-induced calpainactivation and tau toxicity [204]. Calpain is a calcium-dependent cysteine protease responsible for the generation ofthe neurotoxic 17 kDa tau [205, 206]. Decreasing membranecholesterol in mature neurons reduced their susceptibility tothe Aš›½-induced calpain activation, 17 kDa tau production,and cell death, whereas elevated membrane cholesterol lev-els enhanced this Aš›½-triggered cascade in young neurons[204]. Like calpain, AMP-activated protein kinase (AMPK),a serine/threonine kinase, is also activated by elevated cal-cium levels. Interestingly, in addition to its important func-tion in regulating cholesterol homeostasis, emerging studies

  • BioMed Research International 9

    revealed AMPK as a potential tau phosphorylating enzyme[207, 208]. AMPK-induced abnormal tau phosphorylationinhibited microtubule binding of tau [207]. In line withthese findings Vingtdeux et al. reported an accumulation ofactivated AMPK in cerebral neurons of AD brains [209].Other authors, however, even ascribed AMPK an inhibitingfunction in tau phosphorylation by downregulating glycogensynthase kinase-3š›½ (GSK3š›½) activity, one of the main tauphosphorylating kinases [210, 211]. Beside cholesterol, šœ”-3fatty acids are also discussed to influence tau pathology. In a3xTg-AD mouse model, Green et al. demonstrated loweredlevels of intraneuronal tau and reduced tau phosphoryla-tion after 3 to 9 months of DHA supplementation [112].Similar results were also observed by Ma et al. after fishoil administration to 3xTg-AD mice [212]. In both studies,the reduced phosphorylation was attributed to an inhibitionof the c-Jun N-terminal kinase (JNK). In contrast, low šœ”3intake with a decreased šœ”3 :šœ”6 ratio leads to an aggravationof tau pathology in these transgenic mice [213]. Further-more, some studies revealed a colocalization of sphingolipidsand gangliosides with PHFs proposing a possible relationbetween sphingolipid metabolism and tau [214, 215]. Indeed,inhibition of the serine palmitoyl transferase (SPT), therate-limiting enzyme in sphingolipid synthesis, and the firststep towards ceramide synthesis resulted in a reduced tauhyperphosphorylation in an AD mouse model [216]. Aftertreatment of differentiated PC12 cells with ceramide derivates(N-acetylsphingosine, and N-hexanoylsphingosine), Xie andJohnson reported a significant reduction in tau levels withoutaffecting tau phosphorylation. This was attributed to anincreased expression of calpain I and thus stimulated tauprotein degradation [217]. Taken together, not only APPprocessing but also tau pathology is influenced by lipids.Although the general view for APP processing seems moreconsistent, further investigations linking tau to altered lipidhomeostasis should follow.

    6. Conclusion

    Summing it up, lipids are tightly linked to AD. It has beenshown that cholesterol increases amyloidogenic pathwaysand decreases non amyloidogenic pathways followed by anenhanced Aš›½ production and aggregation. Opposite effectswere observed for DHA, suggesting a potential beneficialrole for DHA or PUFAs in AD. For sphingolipids and gly-cosphingolipids, a more complex situation in respect to ADis reported. Although lipids like SM, S1P, and sulfatides seemto be protective by enhancing Aš›½ clearance or decreasingAš›½ production, other glycosphingolipids like gangliosides orceramides increase Aš›½ toxicity or Aš›½ oligomerization. Inter-estingly, it has been shown that, in return, APP processingalso affects lipid metabolism, resulting in complex regulatoryfeed-back cycles, which seem to be dysregulated in AD.In line, several studies suggest an altered lipid metabolismin human AD brains. However, controversial effects arereported in different brain regions and tissues, making moredetailed analysis with new lipidomic approaches and highernumbers necessary.

    References

    [1] T. Dyrks, A. Weidemann, G. Multhaup et al., ā€œIdentification,transmembrane orientation and biogenesis of the amyloid A4precursor of Alzheimerā€™s disease,ā€ EMBO Journal, vol. 7, no. 4,pp. 949ā€“957, 1988.

    [2] B. Anliker and U. MuĢˆller, ā€œThe functions of mammalianamyloid precursor protein and related amyloid precursor-likeproteins,ā€ Neurodegenerative Diseases, vol. 3, no. 4-5, pp. 239ā€“246, 2006.

    [3] J. Herms, B. Anliker, S. Heber et al., ā€œCortical dysplasia resem-bling human type 2 lissencephaly in mice lacking all three APPfamily members,ā€ EMBO Journal, vol. 23, no. 20, pp. 4106ā€“4115,2004.

    [4] F. Magara, U. MuĢˆller, Z. W. Li et al., ā€œGenetic backgroundchanges the pattern of forebrain commissure defects in trans-genic mice underexpressing the š›½-amyloid-precursor protein,ā€Proceedings of the National Academy of Sciences of the UnitedStates of America, vol. 96, no. 8, pp. 4656ā€“4661, 1999.

    [5] J. P. Steinbach, U. MuĢˆller, M. Leist, Z. W. Li, P. Nicotera, andA. Aguzzi, ā€œHypersensitivity to seizures in š›½-amyloid precursorprotein deficientmice,ā€Cell Death andDifferentiation, vol. 5, no.10, pp. 858ā€“866, 1998.

    [6] Tyan, S. -H, Shih et al., ā€œAmyloid precursor protein (APP) reg-ulates synaptic structure and function,ā€ Molecular and CellularNeuroscience, vol. 51, no. 1-2, pp. 43ā€“52, 2012.

    [7] K. J. Lee, C. E. H. Moussa, Y. Lee et al., ā€œBeta amyloid-independent role of amyloid precursor protein in generationandmaintenance of dendritic spines,ā€Neuroscience, vol. 169, no.1, pp. 344ā€“356, 2010.

    [8] H. S. Hoe, Z. Fu, A. Makarova et al., ā€œThe effects of amyloidprecursor protein on postsynaptic composition and activity,ā€Journal of Biological Chemistry, vol. 284, no. 13, pp. 8495ā€“8506,2009.

    [9] F. S. Esch, P. S. Keim, E. C. Beattie et al., ā€œCleavage of amyloid š›½peptide during constitutive processing of its precursor,ā€ Science,vol. 248, no. 4959, pp. 1122ā€“1124, 1990.

    [10] M. P. Mattson, Z. H. Guo, and J. D. Geiger, ā€œSecreted formof amyloid precursor protein enhances basal glucose andglutamate transport and protects against oxidative impairmentof glucose and glutamate transport in synaptosomes by a cyclicGMP-mediatedmechanism,ā€ Journal of Neurochemistry, vol. 73,no. 2, pp. 532ā€“537, 1999.

    [11] H. Meziane, J. C. Dodart, C. Mathis et al., ā€œMemory-enhancingeffects of secreted forms of the š›½-amyloid precursor proteinin normal and amnestic mice,ā€ Proceedings of the NationalAcademy of Sciences of the United States of America, vol. 95, no.21, pp. 12683ā€“12688, 1998.

    [12] S. Ring, S. W.Weyer, S. B. Kilian et al., ā€œThe secreted š›½-amyloidprecursor protein ectodomain APPsš›¼ is sufficient to rescue theanatomical, behavioral, and electrophysiological abnormalitiesof APP-deficient mice,ā€ Journal of Neuroscience, vol. 27, no. 29,pp. 7817ā€“7826, 2007.

    [13] M. Asai, C. Hattori, B. SzaboĢ et al., ā€œPutative function ofADAM9, ADAM10, and ADAM17 as APP š›¼-secretase,ā€ Bio-chemical and Biophysical Research Communications, vol. 301, no.1, pp. 231ā€“235, 2003.

    [14] J. D. Buxbaum, K. N. Liu, Y. Luo et al., ā€œEvidence that tumornecrosis factor š›¼ converting enzyme is involved in regulated š›¼-secretase cleavage of the Alzheimer amyloid protein precursor,ā€Journal of Biological Chemistry, vol. 273, no. 43, pp. 27765ā€“27767, 1998.

  • 10 BioMed Research International

    [15] S. Lammich, E. Kojro, R. Postina et al., ā€œConstitutive and reg-ulated š›¼-secretase cleavage of Alzheimerā€™s amyloid precursorprotein by a disintegrin metalloprotease,ā€ Proceedings of theNational Academy of Sciences of the United States of America,vol. 96, no. 7, pp. 3922ā€“3927, 1999.

    [16] R. Yan, M. J. Blenkowski, M. E. Shuck et al., ā€œMembrane-anchored aspartyl proteasewithAlzheimerā€™s diseaseš›½-secretaseactivity,ā€ Nature, vol. 402, no. 6761, pp. 533ā€“537, 1999.

    [17] R. Vassar, B. D. Bennett, S. Babu-Khan et al., ā€œš›½-secretasecleavage of Alzheimerā€™s amyloid precursor protein by thetransmembrane aspartic protease BACE,ā€ Science, vol. 286, no.5440, pp. 735ā€“741, 1999.

    [18] V. Y. H. Hook, T. Toneff, W. Aaron, S. Yasothornsrikul, R.Bundey, and T. Reisine, ā€œš›½-amyloid peptide in regulated secre-tory vesicles of chromaffin cells: evidence for multiple cysteineproteolytic activities in distinct pathways for š›½-secretase activ-ity in chromaffin vesicles,ā€ Journal of Neurochemistry, vol. 81, no.2, pp. 237ā€“256, 2002.

    [19] X. Hu, C. W. Hicks, W. He et al., ā€œBace1 modulates myelinationin the central and peripheral nervous system,ā€ Nature Neuro-science, vol. 9, no. 12, pp. 1520ā€“1525, 2006.

    [20] M.Willem, A. N. Garratt, B. Novak et al., ā€œControl of peripheralnerve myelination by the š›½-secretase BACE1,ā€ Science, vol. 314,no. 5799, pp. 664ā€“666, 2006.

    [21] W. T. Kimberly, M. J. LaVoie, B. L. Ostaszewski, W. Ye, M. S.Wolfe, and D. J. Selkoe, ā€œš›¾-secretase is a membrane proteincomplex comprised of presenilin, nicastrin, aph-1, and pen-2,ā€Proceedings of the National Academy of Sciences of the UnitedStates of America, vol. 100, no. 11, pp. 6382ā€“6387, 2003.

    [22] B. de Strooper, P. Saftig, K. Craessaerts et al., ā€œDeficiency ofpresenilin-1 inhibits the normal cleavage of amyloid precursorprotein,ā€ Nature, vol. 391, no. 6665, pp. 387ā€“390, 1998.

    [23] M. O. Grimm, I. Tomic, and T. Hartmann, ā€œPotential externalsource of Aš›½ in biological samples,ā€ Nature Cell Biology, vol. 4,pp. E164ā€“E165, 2002.

    [24] E. Levy-Lahad, W. Wasco, P. Poorkaj et al., ā€œCandidate gene forthe chromosome 1 familial Alzheimerā€™s disease locus,ā€ Science,vol. 269, no. 5226, pp. 973ā€“977, 1995.

    [25] B. Grziwa, M. O. W. Grimm, C. L. Masters, K. Beyreuther,T. Hartmann, and S. F. Lichtenthaler, ā€œThe transmembranedomain of the amyloid precursor protein in microsomal mem-branes is on both sides shorter than predicted,ā€ Journal ofBiological Chemistry, vol. 278, no. 9, pp. 6803ā€“6808, 2003.

    [26] G. G. Glenner and C. W. Wong, ā€œAlzheimerā€™s disease: initialreport of the purification and characterization of a novelcerebrovascular amyloid protein,ā€ Biochemical and BiophysicalResearch Communications, vol. 120, no. 3, pp. 885ā€“890, 1984.

    [27] J. T. Jarrett, E. P. Berger, and P. T. Lansbury Jr., ā€œThe carboxyterminus of the š›½ amyloid protein is critical for the seedingof amyloid formation: implications for the pathogenesis ofAlzheimerā€™s disease,ā€ Biochemistry, vol. 32, no. 18, pp. 4693ā€“4697, 1993.

    [28] X. Cao and T. C. Sudhof, ā€œA Transcriptively active complex ofAPP with Fe65 and histone acetyltransferase Tip60,ā€ Science,vol. 293, no. 5527, pp. 115ā€“120, 2001.

    [29] S. Wang, R. Wang, L. Chen, D. A. Bennett, D. W. Dickson, andD. S. Wang, ā€œExpression and functional profiling of neprilysin,insulin-degrading enzyme, and endothelin-converting enzymein prospectively studied elderly and Alzheimerā€™s brain,ā€ Journalof Neurochemistry, vol. 115, no. 1, pp. 47ā€“57, 2010.

    [30] J. Y. Huang, D. M. Hafez, B. D. James, D. A. Bennett, and R. A.Marr, ā€œAltered NEP2 expression and activity in mild cognitive

    impairment and Alzheimerā€™s disease,ā€ Journal of Alzheimerā€™sDisease, vol. 28, no. 2, pp. 433ā€“441, 2012.

    [31] L. C. Walker, M. I. Diamond, K. E. Duff, and B. T. Hyman,ā€œMechanisms of protein seeding in neurodegenerative diseases,ā€JAMA Neurology, vol. 70, no. 3, pp. 304ā€“310, 2013.

    [32] D. Harold, R. Abraham, P. Hollingworth et al., ā€œGenome-wide association study identifies variants at CLU and PICALMassociated with Alzheimerā€™s disease,ā€ Nature Genetics, vol. 41,no. 10, pp. 1088ā€“1093, 2009.

    [33] R. Ferrari, J. H. Moreno, A. T. Minhajuddin et al., ā€œImplicationof common and disease specific variants in CLU, CR1, andPICALM,ā€ Neurobiology of Aging, vol. 33, no. 8, pp. 1846.e7ā€“1846.e18, 2012.

    [34] C. Reitz, G. Jun, A. Naj et al., ā€œVariants in the ATP-bindingcassette transporter (ABCA7), apolipoprotein E šœ–4, and the riskof late-onset Alzheimer disease in African Americans,ā€ Journalof the American Medical Association, vol. 309, no. 14, pp. 1483ā€“1492, 2013.

    [35] T. Nuutinen, T. Suuronen, A. Kauppinen, and A. Salminen,ā€œClusterin: a forgotten player in Alzheimerā€™s disease,ā€ BrainResearch Reviews, vol. 61, no. 2, pp. 89ā€“104, 2009.

    [36] E. M. C. Schrijvers, P. J. Koudstaal, A. Hofman, and M. M. B.Breteler, ā€œPlasma clusterin and the risk of Alzheimer disease,ā€Journal of the AmericanMedical Association, vol. 305, no. 13, pp.1322ā€“1326, 2011.

    [37] P. C. May, M. Lampert-Etchells, S. A. Johnson, J. Poirier, J. N.Masters, and C. E. Finch, ā€œDynamics of gene expression for ahippocampal glycoprotein elevated in Alzheimerā€™s disease andin response to experimental lesions in rat,ā€Neuron, vol. 5, no. 6,pp. 831ā€“839, 1990.

    [38] G. Falgarone and G. Chiocchia, ā€œChapter 8: clusterin: a multi-facet protein at the crossroad of inflammation and autoimmu-nity,ā€ Advances in Cancer Research, vol. 104, pp. 139ā€“170, 2009.

    [39] H. Zhang, J. K. Kim, C. A. Edwards, Z. Xu, R. Taichman, andC. Y. Wang, ā€œClusterin inhibits apoptosis by interacting withactivated Bax,ā€ Nature Cell Biology, vol. 7, no. 9, pp. 909ā€“915,2005.

    [40] J. J. Yerbury, S. Poon, S. Meehan et al., ā€œThe extracellularchaperone clusterin influences amyloid formation and toxicityby interacting with prefibrillar structures,ā€ FASEB Journal, vol.21, no. 10, pp. 2312ā€“2322, 2007.

    [41] W. S. Kim, M. L. Fitzgerald, K. Kang et al., ā€œAbca7 null miceretain normalmacrophage phosphatidylcholine and cholesterolefflux activity despite alterations in adipose mass and serumcholesterol levels,ā€ Journal of Biological Chemistry, vol. 280, no.5, pp. 3989ā€“3995, 2005.

    [42] Y. Ikeda, S. Abe-Dohmae, Y. Munehira et al., ā€œPosttranscrip-tional regulation of human ABCA7 and its function for theapoA-I-dependent lipid release,ā€ Biochemical and BiophysicalResearch Communications, vol. 311, no. 2, pp. 313ā€“318, 2003.

    [43] S. L. Chan, W. S. Kim, J. B. Kwok et al., ā€œATP-binding cassettetransporter A7 regulates processing of amyloid precursor pro-tein in vitro,ā€ Journal of Neurochemistry, vol. 106, no. 2, pp. 793ā€“804, 2008.

    [44] M.Kivipelto, E. L.Helkala,M. P. Laakso et al., ā€œApolipoprotein Ešœ€4 allele, elevatedmidlife total cholesterol level, andhighmidlifesystolic blood pressure are independent risk factors for late-lifeAlzheimer disease,ā€ Annals of Internal Medicine, vol. 137, no. 3,pp. 149ā€“155, 2002.

    [45] M. A. Pappolla, T. K. Bryant-Thomas, D. Herbert et al., ā€œMildhypercholesterolemia is an early risk factor for the development

  • BioMed Research International 11

    of Alzheimer amyloid pathology,ā€ Neurology, vol. 61, no. 2, pp.199ā€“205, 2003.

    [46] A. Solomon, M. Kivipelto, B. Wolozin, J. Zhou, and R. A.Whitmer, ā€œMidlife serum cholesterol and increased risk ofAlzheimerā€™s and vascular dementia three decades later,ā€Demen-tia and Geriatric Cognitive Disorders, vol. 28, no. 1, pp. 75ā€“80,2009.

    [47] D. LuĢˆtjohann, A. Papassotiropoulos, I. BjoĢˆrkhem et al.,ā€œPlasma 24S-hydroxycholesterol (cerebrosterol) is increased inAlzheimer and vascular demented patients,ā€ Journal of LipidResearch, vol. 41, no. 2, pp. 195ā€“198, 2000.

    [48] J. Y. Hur, H. Welander, H. Behbahani et al., ā€œActive š›¾-secretaseis localized to detergent-resistant membranes in human brain,ā€FEBS Journal, vol. 275, no. 6, pp. 1174ā€“1187, 2008.

    [49] J. M. Cordy, I. Hussain, C. Dingwall, N. M. Hooper, and A. J.Turner, ā€œExclusively targeting š›½-secretase to lipid rafts by GPI-anchor addition up-regulates š›½-site processing of the amyloidprecursor protein,ā€ Proceedings of the National Academy ofSciences of the United States of America, vol. 100, no. 20, pp.11735ā€“11740, 2003.

    [50] E. Kojro, G. Gimpl, S. Lammich, W. MaĢˆrz, and F. Fahrenholz,ā€œLow cholesterol stimulates the nonamyloidogenic pathway byits effect on the š›¼-secretase ADAM 10,ā€ Proceedings of theNational Academy of Sciences of the United States of America,vol. 98, no. 10, pp. 5815ā€“5820, 2001.

    [51] A. J. Beel, M. Sakakura, P. J. Barrett, and C. R. Sanders,ā€œDirect binding of cholesterol to the amyloid precursor protein:an important interaction in lipid-Alzheimerā€™s disease relation-ships?ā€ Biochimica et Biophysica Acta, vol. 1801, no. 8, pp. 975ā€“982, 2010.

    [52] P. J. Barrett, Y. Song, W. D. van Horn et al., ā€œThe amyloidprecursor protein has a flexible transmembrane domain andbinds cholesterol,ā€ Science, vol. 336, no. 6085, pp. 1168ā€“1171, 2012.

    [53] M. O. W. Grimm, H. S. Grimm, I. Tomic, K. Beyreuther,T. Hartmann, and C. Bergmann, ā€œIndependent inhibition ofAlzheimer disease š›½- and š›¾-secretase cleavage by loweredcholesterol levels,ā€ Journal of Biological Chemistry, vol. 283, no.17, pp. 11302ā€“11311, 2008.

    [54] P. Osenkowski, W. Ye, R. Wang, M. S. Wolfe, and D. J.Selkoe, ā€œDirect and potent regulation of š›¾-secretase by its lipidmicroenvironment,ā€ Journal of Biological Chemistry, vol. 283,no. 33, pp. 22529ā€“22540, 2008.

    [55] L. Kalvodova, N. Kahya, P. Schwille et al., ā€œLipids as modulatorsof proteolytic activity of BACE: involvement of cholesterol,glycosphingolipids, and anionic phospholipids in vitro,ā€ Journalof Biological Chemistry, vol. 280, no. 44, pp. 36815ā€“36823, 2005.

    [56] A. Hosaka, W. Araki, A. Oda, Y. Tomidokoro, and A. Tamaoka,ā€œStatins reduce amyloid š›½-peptide production by modulatingamyloid precursor protein maturation and phosphorylationthrough a cholesterol-independent mechanism in culturedneurons,ā€ Neurochemical Research, vol. 38, no. 3, pp. 589ā€“600,2013.

    [57] E.Winkler, F. Kamp, J. Scheuring, A. Ebke, A. Fukumori, andH.Steiner, ā€œGeneration of Alzheimer disease-associated amyloidbeta42/43 peptide by gamma-secretase can be inhibited directlyby modulation of membrane thickness,ā€ Journal of BiologicalChemistry, vol. 287, no. 25, pp. 21326ā€“21334, 2012.

    [58] C. Haass, M. G. Schlossmacher, A. Y. Hung et al., ā€œAmy-loid š›½-peptide is produced by cultured cells during normalmetabolism,ā€ Nature, vol. 359, no. 6393, pp. 322ā€“325, 1992.

    [59] K. N. Dahlgren, A. M. Manelli, W. Blaine Stine Jr., L. K. Baker,G. A. Krafft, and M. J. Ladu, ā€œOligomeric and fibrillar species

    of amyloid-š›½ peptides differentially affect neuronal viability,ā€Journal of Biological Chemistry, vol. 277, no. 35, pp. 32046ā€“32053, 2002.

    [60] A. Schneider, W. Schulz-Schaeffer, T. Hartmann, J. B. Schulz,and M. Simons, ā€œCholesterol depletion reduces aggregation ofamyloid-beta peptide in hippocampal neurons,ā€Neurobiology ofDisease, vol. 23, no. 3, pp. 573ā€“577, 2006.

    [61] X. Zhou and J. Xu, ā€œFree cholesterol induces higher beta-sheetcontent inAbeta peptide oligomers by aromatic interactionwithPhe19,ā€ PLoS ONE, vol. 7, no. 9, Article ID e46245, 2012.

    [62] A. Y. Abramov, M. Ionov, E. Pavlov, and M. R. Duchen, ā€œMem-brane cholesterol content plays a key role in the neurotoxicityof š›½-amyloid: implications for Alzheimerā€™s disease,ā€ Aging Cell,vol. 10, no. 4, pp. 595ā€“603, 2011.

    [63] P. Ferrera, O. Mercado-GoĢmez, M. Silva-Aguilar, M. Valverde,and C. Arias, ā€œCholesterol potentiates š›½-amyloid-induced tox-icity in human neuroblastoma cells: involvement of oxidativestress,ā€ Neurochemical Research, vol. 33, no. 8, pp. 1509ā€“1517,2008.

    [64] I. Sponne, A. Fifre, V. Koziel, T. Oster, J. L. Olivier, and T. Pil-lot, ā€œMembrane cholesterol interferes with neuronal apoptosisinduced by soluble oligomers but not fibrils of amyloid-betapeptide,ā€The FASEB Journal, vol. 18, no. 7, pp. 836ā€“838, 2004.

    [65] N. Arispe andM. Doh, ā€œPlasma membrane cholesterol controlsthe cytotoxicity of Alzheimerā€™s disease Aš›½P (1-40) and (1-42)peptides,ā€ FASEB Journal, vol. 16, no. 12, pp. 1526ā€“1536, 2002.

    [66] M. O. W. Grimm, H. S. Grimm, A. J. PaĢˆtzold et al., ā€œRegulationof cholesterol and sphingomyelinmetabolismby amyloid-š›½ andpresenilin,ā€Nature Cell Biology, vol. 7, no. 11, pp. 1118ā€“1123, 2005.

    [67] E. Canepa, R. Borghi, J. Via et al., ā€œCholesterol and amyloid-š›½:evidence for a cross-talk between astrocytes and neuronal cells,ā€Journal of Alzheimerā€™s Disease, vol. 25, no. 4, pp. 645ā€“653, 2011.

    [68] M. O. W. Grimm, J. A. TschaĢˆpe, H. S. Grimm, E. G. Zinser,and T. Hartmann, ā€œAltered membrane fluidity and lipid raftcomposition in presenilin-deficient cells,ā€ Acta NeurologicaScandinavica, vol. 114, no. 185, pp. 27ā€“32, 2006.

    [69] T. Umeda, H. Mori, H. Zheng, and T. Tomiyama, ā€œRegulationof cholesterol efflux by amyloid š›½ secretion,ā€ Journal of Neuro-science Research, vol. 88, no. 9, pp. 1985ā€“1994, 2010.

    [70] T. J. L. van de Parre, P. J. D. F. Guns, P. Fransen et al.,ā€œAttenuated atherogenesis in apolipoprotein E-deficient micelacking amyloid precursor protein,ā€ Atherosclerosis, vol. 216, no.1, pp. 54ā€“58, 2011.

    [71] A. Nunes, S. N. R. Pressey, J. D. Cooper, and S. Soriano, ā€œLossof amyloid precursor protein in a mouse model of Niemann-Pick type C disease exacerbates its phenotype and disrupts tauhomeostasis,ā€ Neurobiology of Disease, vol. 42, no. 3, pp. 349ā€“359, 2011.

    [72] Q. Liu, C. V. Zerbinatti, J. Zhang et al., ā€œAmyloid precur-sor protein regulates brain apolipoprotein E and cholesterolmetabolism through lipoprotein receptor LRP1,ā€ Neuron, vol.56, no. 1, pp. 66ā€“78, 2007.

    [73] M. Hashimoto, H. M. Shahdat, S. Yamashita et al., ā€œDocosa-hexaenoic acid disrupts in vitro amyloid š›½1-40 fibrillation andconcomitantly inhibits amyloid levels in cerebral cortex ofAlzheimerā€™s disease model rats,ā€ Journal of Neurochemistry, vol.107, no. 6, pp. 1634ā€“1646, 2008.

    [74] M. O. W. Grimm, J. Kuchenbecker, S. Grosgen et al., ā€œDocosa-hexaenoic acid reduces amyloid š›½ production via multiplepleiotropic mechanisms,ā€ Journal of Biological Chemistry, vol.286, no. 16, pp. 14028ā€“14039, 2011.

  • 12 BioMed Research International

    [75] Y. Zhao, F. Calon, C. Julien et al., ā€œDocosahexaenoic acid-derived neuroprotectin D1 induces neuronal survival viasecretase- and PPARš›¾-mediated mechanisms in Alzheimerā€™sdisease models,ā€ PLoS ONE, vol. 6, no. 1, Article ID e15816, 2011.

    [76] W. J. Lukiw, J. G. Cui, V. L. Marcheselli et al., ā€œA role fordocosahexaenoic acid-derived neuroprotectin D1 in neural cellsurvival and Alzheimer disease,ā€ Journal of Clinical Investiga-tion, vol. 115, no. 10, pp. 2774ā€“2783, 2005.

    [77] E. Hjorth, M. Zhu, V. C. Toro et al., ā€œOmega-3 Fatty AcidsEnhance Phagocytosis of Alzheimerā€™s Disease-Related Am-yloid-beta42 by Human Microglia and Decrease InflammatoryMarkers,ā€ Journal of Alzheimerā€™s Disease, vol. 35, no. 4, pp. 697ā€“713, 2013.

    [78] V. F. Labrousse, A. Nadjar, C. Joffre et al., ā€œShort-term longchain omega3 diet protects from neuroinflammatory processesand memory impairment in aged mice,ā€ PLoS One, vol. 7, no. 5,Article ID e36861, 2012.

    [79] Q. L. Ma, B. Teter, O. J. Ubeda et al., ā€œOmega-3 fatty aciddocosahexaenoic acid increases SorLA/LR11, a sorting proteinwith reduced expression in sporadic Alzheimerā€™s disease (AD):relevance toADprevention,ā€ Journal of Neuroscience, vol. 27, no.52, pp. 14299ā€“14307, 2007.

    [80] D. L. Sparks, S. W. Scheff, J. C. Hunsaker III, H. Liu, T.Landers, and D. R. Gross, ā€œInduction of Alzheimer-like š›½-amyloid immunoreactivity in the brains of rabbits with dietarycholesterol,ā€ Experimental Neurology, vol. 126, no. 1, pp. 88ā€“94,1994.

    [81] F. S. Shie, L. W. Jin, D. G. Cook, J. B. Leverenz, and R. C.LeBoeuf, ā€œDiet-induced hypercholesterolemia enhances brainAš›½ accumulation in transgenic mice,ā€ NeuroReport, vol. 13, no.4, pp. 455ā€“459, 2002.

    [82] L. M. Refolo, B. Malester, J. LaFrancois et al., ā€œHypercholes-terolemia accelerates the Alzheimerā€™s amyloid pathology in atransgenic mouse model,ā€ Neurobiology of Disease, vol. 7, no. 4,pp. 321ā€“331, 2000.

    [83] L. M. Refolo, M. A. Pappolla, J. LaFrancois et al., ā€œA cholesterol-lowering drug reduces š›½-amyloid pathology in a transgenicmouse model of Alzheimerā€™s disease,ā€ Neurobiology of Disease,vol. 8, no. 5, pp. 890ā€“899, 2001.

    [84] T. Kurata, K. Miyazaki, M. Kozuki et al., ā€œAtorvastatin andpitavastatin reduce senile plaques and inflammatory responsesin a mouse model of Alzheimerā€™s disease,ā€ Journal of NeurologyResearch, vol. 34, no. 6, pp. 601ā€“610, 2012.

    [85] N. Sato, M. Shinohara, H. Rakugi, and R. Morishita, ā€œDualeffects of statins on Aš›½ metabolism: upregulation of thedegradation of APP-CTF and Aš›½ clearance,ā€NeurodegenerativeDiseases, vol. 10, no. 1ā€“4, pp. 305ā€“308, 2012.

    [86] K. Fassbender, M. Simons, C. Bergmann et al., ā€œSimvastatinstrongly reduces levels of Alzheimerā€™s disease š›½-amyloid pep-tides Aš›½42 and Aš›½40 in vitro and in vivo,ā€ Proceedings of theNational Academy of Sciences of the United States of America,vol. 98, no. 10, pp. 5856ā€“5861, 2001.

    [87] L. Cibickova, H. Radomir, M. Stanislav et al., ā€œThe influenceof simvastatin, atorvastatin and high-cholesterol diet on acetyl-cholinesterase activity, amyloid beta and cholesterol synthesisin rat brain,ā€ Steroids, vol. 74, no. 1, pp. 13ā€“19, 2009.

    [88] I. H. Park, E. M. Hwang, H. S. Hong et al., ā€œLovastatin enhancesAš›½ production and senile plaque deposition in female Tg2576mice,ā€ Neurobiology of Aging, vol. 24, no. 5, pp. 637ā€“643, 2003.

    [89] M. D. M. Haag, A. Hofman, P. J. Koudstaal, B. H. C. Stricker,andM.M. B. Breteler, ā€œStatins are associated with a reduced risk

    of Alzheimer disease regardless of lipophilicity. The RotterdamStudy,ā€ Journal of Neurology, Neurosurgery and Psychiatry, vol.80, no. 1, pp. 13ā€“17, 2009.

    [90] B. Wolozin, W. Kellman, P. Ruosseau, G. G. Celesia, and G.Siegel, ā€œDecreased prevalence of Alzheimer disease associ-ated with 3-hydroxy-3-methyglutaryl coenzyme A reductaseinhibitors,ā€ Archives of Neurology, vol. 57, no. 10, pp. 1439ā€“1443,2000.

    [91] B. Wolozin, S. W. Wang, N. C. Li, A. Lee, T. A. Lee, and L. E.Kazis, ā€œSimvastatin is associated with a reduced incidence ofdementia and Parkinsonā€™s disease,ā€ BMCMedicine, vol. 5, article20, 2007.

    [92] T. D. Rea, J. C. Breitner, B. M. Psaty et al., ā€œStatin use and therisk of incident dementia: the Cardiovascular Health Study,ā€Archives of Neurology, vol. 62, no. 7, pp. 1047ā€“1051, 2005.

    [93] Z. Arvanitakis, J. A. Schneider, R. S. Wilson et al., ā€œStatins,incident Alzheimer disease, change in cognitive function, andneuropathology,ā€Neurology, vol. 70, no. 19, pp. 1795ā€“1802, 2008.

    [94] H. H. Feldman, R. S. Doody, M. Kivipelto et al., ā€œRandomizedcontrolled trial of atorvastatin in mild to moderate Alzheimerdisease: LEADe,ā€ Neurology, vol. 74, no. 12, pp. 956ā€“964, 2010.

    [95] M. Sano, K. L. Bell, D. Galasko et al., ā€œA randomized, double-blind, placebo-controlled trial of simvastatin to treat Alzheimerdisease,ā€ Neurology, vol. 77, no. 6, pp. 556ā€“563, 2011.

    [96] R. J. Pawlosky, J. R. Hibbeln, J. A. Novotny, and N. SalemJr., ā€œPhysiological compartmental analysis of š›¼-linolenic acidmetabolism in adult humans,ā€ Journal of Lipid Research, vol. 42,no. 8, pp. 1257ā€“1265, 2001.

    [97] S. R. Wassall and W. Stillwell, ā€œPolyunsaturated fatty acid-cholesterol interactions: domain formation in membranes,ā€Biochimica et Biophysica Acta, vol. 1788, no. 1, pp. 24ā€“32, 2009.

    [98] S. Gamoh, M. Hashimoto, K. Sugioka et al., ā€œChronic admin-istration of docosahexaenoic acid improves reference memory-related learning ability in young rats,ā€ Neuroscience, vol. 93, no.1, pp. 237ā€“241, 1999.

    [99] M. Katakura, M. Hashimoto, H. M. Shahdat et al., ā€œDocosa-hexaenoic acid promotes neuronal differentiation by regulatingbasic helix-loop-helix transcription factors and cell cycle inneural stem cells,ā€ Neuroscience, vol. 160, no. 3, pp. 651ā€“660,2009.

    [100] L. Dagai, R. Peri-Naor, and R. Z. Birk, ā€œDocosahexaenoic acidsignificantly stimulates immediate early response genes andneurite outgrowth,ā€ Neurochemical Research, vol. 34, no. 5, pp.867ā€“875, 2009.

    [101] F. Calon, G. P. Lim, F. Yang et al., ā€œDocosahexaenoic acidprotects from dendritic pathology in an Alzheimerā€™s diseasemouse model,ā€ Neuron, vol. 43, no. 5, pp. 633ā€“645, 2004.

    [102] M. Soderberg, C. Edlund, K. Kristensson, and G. Dallner,ā€œFatty acid composition of brain phospholipids in aging and inAlzheimerā€™s disease,ā€ Lipids, vol. 26, no. 6, pp. 421ā€“425, 1991.

    [103] T. J. Montine and J. D. Morrow, ā€œFatty acid oxidation inthe pathogenesis of Alzheimerā€™s disease,ā€ American Journal ofPathology, vol. 166, no. 5, pp. 1283ā€“1289, 2005.

    [104] W. R. Markesbery, R. J. Kryscio, M. A. Lovell, and J. D. Morrow,ā€œLipid peroxidation is an early event in the brain in amnesticmild cognitive impairment,ā€ Annals of Neurology, vol. 58, no. 5,pp. 730ā€“735, 2005.

    [105] S. Kalmijn, L. J. Launer, A. Ott, J. C. M. Witteman, A. Hofman,andM.M. B. Breteler, ā€œDietary fat intake and the risk of incidentdementia in the Rotterdam study,ā€ Annals of Neurology, vol. 42,no. 5, pp. 776ā€“782, 1997.

  • BioMed Research International 13

    [106] M. J. Engelhart, M. I. Geerlings, A. Ruitenberg et al., ā€œDietand risk of dementia: does fat matter? The Rotterdam study,ā€Neurology, vol. 59, no. 12, pp. 1915ā€“1921, 2002.

    [107] P. Barberger-Gateau, L. Letenneur, V. Deschamps, K. PeĢreĢ€s, J.F. Dartigues, and S. Renaud, ā€œFish, meat, and risk of dementia:cohort study,ā€ British Medical Journal, vol. 325, no. 7370, pp.932ā€“933, 2002.

    [108] M. C. Morris, D. A. Evans, J. L. Bienias et al., ā€œConsumption offish and n-3 fatty acids and risk of incident Alzheimer disease,ā€Archives of Neurology, vol. 60, no. 7, pp. 940ā€“946, 2003.

    [109] E. J. Schaefer, V. Bongard, A. S. Beiser et al., ā€œPlasma phos-phatidylcholine docosahexaenoic acid content and risk ofdementia and alzheimer disease: the framingham heart study,ā€Archives of Neurology, vol. 63, no. 11, pp. 1545ā€“1550, 2006.

    [110] L. J. Whalley, I. J. Deary, J. M. Starr et al., ā€œn-3 Fatty acid ery-throcyte membrane content, APOE šœ€4, and cognitive variation:an observational follow-up study in late adulthood,ā€ AmericanJournal of Clinical Nutrition, vol. 87, no. 2, pp. 449ā€“454, 2008.

    [111] S. Oddo, A. Caccamo, J. D. Shepherd et al., ā€œTriple-transgenicmodel of Alzheimerā€™s disease with plaques and tangles: intra-cellular Aš›½ and synaptic dysfunction,ā€Neuron, vol. 39, no. 3, pp.409ā€“421, 2003.

    [112] K. N. Green, H. Martinez-Coria, H. Khashwji et al., ā€œDietarydocosahexaenoic acid and docosapentaenoic acid ameliorateamyloid-š›½ and tau pathology via a mechanism involving prese-nilin 1 levels,ā€ Journal of Neuroscience, vol. 27, no. 16, pp. 4385ā€“4395, 2007.

    [113] G. P. Lim, F. Calon, T. Morihara et al., ā€œA diet enrichedwith the omega-3 fatty acid docosahexaenoic acid reducesamyloid burden in an aged Alzheimer mouse model,ā€ Journalof Neuroscience, vol. 25, no. 12, pp. 3032ā€“3040, 2005.

    [114] M. Hashimoto, S. Hossain, T. Shimada, and O. Shido, ā€œDocosa-hexaenoic acid-induced protective effect against impairedlearning in amyloid š›½-infused rats is associated with increasedsynaptosomal membrane fluidity,ā€ Clinical and ExperimentalPharmacology and Physiology, vol. 33, no. 10, pp. 934ā€“939, 2006.

    [115] P. K. Mukherjee, V. L. Marcheselli, C. N. Serhan, and N. G.Bazan, ā€œNeuroprotectin D1: a docosahexaenoic acid-deriveddocosatriene protects human retinal pigment epithelial cellsfrom oxidative stress,ā€ Proceedings of the National Academy ofSciences of the United States of America, vol. 101, no. 22, pp. 8491ā€“8496, 2004.

    [116] Y. Freund-Levi, M. Eriksdotter-JoĢˆnhagen, T. Cederholm et al.,ā€œšœ”-3 fatty acid treatment in 174 patients with mild to moderateAlzheimer disease: omegAD studyā€”a randomized double-blind trial,ā€ Archives of Neurology, vol. 63, no. 10, pp. 1402ā€“1408,2006.

    [117] S. Kotani, E. Sakaguchi, S. Warashina et al., ā€œDietary supple-mentation of arachidonic and docosahexaenoic acids improvescognitive dysfunction,ā€Neuroscience Research, vol. 56, no. 2, pp.159ā€“164, 2006.

    [118] C. C. Chiu, K. P. Su, T. C. Cheng et al., ā€œThe effects of ome-ga-3 fatty acids monotherapy in Alzheimerā€™s disease andmild cognitive impairment: a preliminary randomizeddouble-blind placebo-controlled study,ā€ Progress in Neuro-Psychopharmacology and Biological Psychiatry, vol. 32, no. 6,pp. 1538ā€“1544, 2008.

    [119] P. Scheltens, P. J. G. H. Kamphuis, F. R. J. Verhey et al., ā€œEfficacyof a medical food in mild Alzheimerā€™s disease: a randomized,controlled trial,ā€ Alzheimerā€™s and Dementia, vol. 6, no. 1, pp. 1ā€“10, 2010.

    [120] P. Scheltens, J.W. Twisk, R. Blesa et al., ā€œEfficacy of Souvenaid inmild Alzheimerā€™s disease: results from a randomized, controlledtrial,ā€ Journal of Alzheimerā€™s Disease, vol. 31, no. 1, pp. 225ā€“236,2012.

    [121] P. Scheltens, ā€œAlzheimerā€™s and Parkinsonā€™s diseases: mecha-nisms, clinical strategies, and promising treatments of neu-rodegenerative diseases,ā€ in Proceedings of the 11th InternationalConference AD/PD, Karger, Florence, Italy, 2013.

    [122] M. O. W. Grimm, S. Grsgen, T. L. Rothhaar et al., ā€œIntracel-lular APP domain regulates serine-palmitoyl-CoA transferaseexpression and is affected in alzheimerā€™s disease,ā€ InternationalJournal of Alzheimerā€™s Disease, vol. 2011, Article ID 695413, 8pages, 2011.

    [123] J. Tarasiuk, K. Kapica-Topczewska, A. Kulakowska et al.,ā€œIncreased concentration of the CSF Tau protein and its phos-phorylated form in the late juvenile metachromatic leukodys-trophy form: a case report,ā€ Journal of Neural Transmission, vol.119, no. 7, pp. 759ā€“762, 2012.

    [124] S. Keilani, Y. Lun, A. C. Stevens et al., ā€œLysosomal dysfunctionin a mouse model of sandhoff disease leads to accumulation ofganglioside-bound amyloid-š›½ peptide,ā€ Journal of Neuroscience,vol. 32, no. 15, pp. 5223ā€“5236, 2012.

    [125] X. Han, D. M. Holtzman, D. W. McKeel Jr., J. Kelley, andJ. C. Morris, ā€œSubstantial sulfatide deficiency and ceramideelevation in very early Alzheimerā€™s disease: potential role indisease pathogenesis,ā€ Journal of Neurochemistry, vol. 82, no. 4,pp. 809ā€“818, 2002.

    [126] T. Taki, ā€œAn approach to glycobiology from glycolipidomics:ganglioside molecular scanning in the brains of patientswith Alzheimerā€™s disease by TLC-blot/matrix assisted laserdesorption/ionization-time of flight MS,ā€ Biological & Pharma-ceutical Bulletin, vol. 35, no. 10, pp. 1642ā€“1647, 2012.

    [127] Z. Pernber, K. Blennow, N. Bogdanovic, J. E. Mansson, and M.Blomqvist, ā€œAltered distribution of the gangliosides GM1 andGM2 in Alzheimerā€™s disease,ā€ Dementia and Geriatric CognitiveDisorders, vol. 33, no. 2-3, pp. 174ā€“188, 2012.

    [128] V. Filippov, M. A. Song, K. Zhang et al., ā€œIncreased ceramide inbrains with alzheimerā€™s and other neurodegenerative diseases,ā€Journal of Alzheimerā€™s Disease, vol. 29, no. 3, pp. 537ā€“547, 2012.

    [129] L. Hejazi, J. W. H.Wong, D. Cheng et al., ā€œMass and relative elu-tion time profiling: two-dimensional analysis of sphingolipidsin Alzheimerā€™s disease brains,ā€ Biochemical Journal, vol. 438, no.1, pp. 165ā€“175, 2011.

    [130] V.MartĢÄ±n, N. Fabelo, G. Santpere et al., ā€œLipid alterations in lipidrafts from Alzheimerā€™s disease human brain cortex,ā€ Journal ofAlzheimerā€™s Disease, vol. 19, no. 2, pp. 489ā€“502, 2010.

    [131] X. He, Y. Huang, B. Li, C. X. Gong, and E. H. Schuchman,ā€œDeregulation of sphingolipid metabolism in Alzheimerā€™s dis-ease,ā€ Neurobiology of Aging, vol. 31, no. 3, pp. 398ā€“408, 2010.

    [132] R. G. Cutler, J. Kelly, K. Storie et al., ā€œInvolvement of oxida-tive stress-induced abnormalities in ceramide and cholesterolmetabolism in brain aging andAlzheimerā€™s disease,ā€Proceedingsof the National Academy of Sciences of the United States ofAmerica, vol. 101, no. 7, pp. 2070ā€“2075, 2004.

    [133] P. Katsel, C. Li, and V. Haroutunian, ā€œGene expression alter-ations in the sphingolipid metabolism pathways during pro-gression of dementia and Alzheimerā€™s disease: a shift towardceramide accumulation at the earliest recognizable stages ofAlzheimerā€™s disease?ā€ Neurochemical Research, vol. 32, no. 4-5,pp. 845ā€“856, 2007.

    [134] M. M. Mielke, V. V. R. Bandaru, N. J. Haughey et al., ā€œSerumceramides increase the risk of Alzheimer disease: the Womenā€™s

  • 14 BioMed Research International

    Health and Aging Study II,ā€ Neurology, vol. 79, no. 7, pp. 633ā€“641, 2012.

    [135] L. Barrier, S. Ingrand, B. Fauconneau, and G. Page, ā€œGender-dependent accumulation of ceramides in the cerebral cortexof the APPSL/PS1Ki mouse model of Alzheimerā€™s disease,ā€Neurobiology of Aging, vol. 31, no. 11, pp. 1843ā€“1853, 2010.

    [136] L. Barrier, B. Fauconneau, A. Nol, and S. Ingrand, ā€œCeramideand related-sphingolipid levels are not altered in disease-associated brain regions of APPSL and APPSL/PS1M146L mousemodels of Alzheimerā€™s disease: relationshipwith the lack of neu-rodegeneration?ā€ International Journal of Alzheimerā€™s Disease,vol. 2011, Article ID 920958, 10 pages, 2011.

    [137] G. Dawson, R. Goswami, J. Kilkus, D. Wiesner, and S. Dawson,ā€œThe formation of ceramide from sphingomyelin is associatedwith cellular apoptosis,ā€Acta Biochimica Polonica, vol. 45, no. 2,pp. 287ā€“297, 1998.

    [138] R. E. Toman, V. Movsesyan, S. K. Murthy, S. Milstien, S.Spiegel, and A. I. Faden, ā€œCeramide-induced cell death inprimary neuronal cultures: upregulation of ceramide levelsduring neuronal apoptosis,ā€ Journal of Neuroscience Research,vol. 68, no. 3, pp. 323ā€“330, 2002.

    [139] X. Zhang, J. Wu, Y. Dou et al., ā€œAsiatic acid protects primaryneurons against C2-ceramide-induced apoptosis,ā€ EuropeanJournal of Pharmacology, vol. 679, no. 1ā€“3, pp. 51ā€“59, 2012.

    [140] J. T. Lee, J. Xu, J.M. Lee et al., ā€œAmyloid-š›½ peptide induces oligo-dendrocyte death by activating the neutral sphingomyelinase-ceramide pathway,ā€ Journal of Cell Biology, vol. 164, no. 1, pp.123ā€“131, 2004.

    [141] E.G. Zinser, T.Hartmann, andM.O.W.Grimm, ā€œAmyloid beta-protein and lipid metabolism,ā€ Biochimica et Biophysica Acta,vol. 1768, no. 8, pp. 1991ā€“2001, 2007.

    [142] G.Wang,M. Dinkins, Q. He et al., ā€œAstrocytes secrete exosomesenriched with proapoptotic ceramide and prostate apoptosisresponse 4 (PAR-4): potential mechanism of apoptosis induc-tion inAlzheimer disease (AD),ā€ Journal of Biological Chemistry,vol. 287, no. 25, pp. 21384ā€“21395, 2012.

    [143] K. Trajkovic, C. Hsu, S. Chiantia et al., ā€œCeramide triggersbudding of exosome vesicles into multivesicular endosomes,ā€Science, vol. 319, no. 5867, pp. 1244ā€“1247, 2008.

    [144] L. Puglielli, B. C. Ellis, A. J. Saunders, and D. M. Kovacs,ā€œCeramide stabilizes š›½-site amyloid precursor protein-cleavingenzyme 1 and promotes amyloid š›½-peptide biogenesis,ā€ Journalof Biological Chemistry, vol. 278, no. 22, pp. 19777ā€“19783, 2003.

    [145] H. K. Mi and L. Puglielli, ā€œTwo Endoplasmic Reticulum(ER)/ER golgi intermediate compartment-based lysine acetyl-transferases post-translationally regulate BACE1 levels,ā€ Journalof Biological Chemistry, vol. 284, no. 4, pp. 2482ā€“2492, 2009.

    [146] L. C. Edsall, G. G. Pirianov, and S. Spiegel, ā€œInvolvementof sphingosine 1-phosphate in nerve growth factor-mediatedneuronal survival and differentiation,ā€ Journal of Neuroscience,vol. 17, no. 18, pp. 6952ā€“6960, 1997.

    [147] R.A. Rius, L. C. Edsall, and S. Spiegel, ā€œActivation of sphingosinekinase in pheochromocytoma PC12 neuronal cells in responseto trophic factors,ā€ FEBS Letters, vol. 417, no. 2, pp. 173ā€“176, 1997.

    [148] N. Takasugi, T. Sasaki, K. Suzuki et al., ā€œBACE1 activity ismodulated by cell-associated sphingosine-1-phosphate,ā€ Journalof Neuroscience, vol. 31, no. 18, pp. 6850ā€“6857, 2011.

    [149] O. Ben-David and A. H. Futerman, ā€œThe role of the ceramideacyl chain length in neurodegeneration: involvement ofceramide synthases,ā€ NeuroMolecular Medicine, vol. 12, no. 4,pp. 341ā€“350, 2010.

    [150] V. V. R. Bandaru, J. Troncoso, D.Wheeler et al., ā€œApoE4 disruptssterol and sphingolipid metabolism in Alzheimerā€™s but notnormal brain,ā€Neurobiology of Aging, vol. 30, no. 4, pp. 591ā€“599,2009.

    [151] J. W. Pettegrew, K. Panchalingam, R. L. Hamilton, and R.J. Mcclure, ā€œBrain membrane phospholipid alterations inAlzheimerā€™s disease,ā€ Neurochemical Research, vol. 26, no. 7, pp.771ā€“782, 2001.

    [152] M. Kosicek, H. Zetterberg, N. Andreasen, J. Peter-Katalinic,and S. Hecimovic, ā€œElevated cerebrospinal fluid sphingomyelinlevels in prodromal Alzheimerā€™s disease,ā€ Neuroscience Letters,vol. 516, no. 2, pp. 302ā€“305, 2012.

    [153] M. M. Mielke, N. J. Haughey, V. V. R. Bandaru et al., ā€œPlasmasphingomyelins are associated with cognitive progression inalzheimerā€™s disease,ā€ Journal of Alzheimerā€™s Disease, vol. 27, no.2, pp. 259ā€“269, 2011.

    [154] K. A. Sheikh, J. Sun, Y. Liu et al., ā€œMice lacking complexgangliosides develop Wallerian degeneration and myelinationdefects,ā€ Proceedings of the National Academy of Sciences of theUnited States of America, vol. 96, no. 13, pp. 7532ā€“7537, 1999.

    [155] T. Yamashita, R.Wada, T. Sasaki et al., ā€œA vital role for glycosph-ingolipid synthesis during development and differentiation,ā€Proceedings of the National Academy of Sciences of the UnitedStates of America, vol. 96, no. 16, pp. 9142ā€“9147, 1999.

    [156] S. Lahiri and A. H. Futerman, ā€œThemetabolism and function ofsphingolipids and glycosphingolipids,ā€ Cellular and MolecularLife Sciences, vol. 64, no. 17, pp. 2270ā€“2284, 2007.

    [157] K. Simons and E. Ikonen, ā€œFunctional rafts in cell membranes,ā€Nature, vol. 387, no. 6633, pp. 569ā€“572, 1997.

    [158] I. Kracun, H. Rosner, V. Drnovsek, M. Heffer-Lauc, C. Cosovic,and G. Lauc, ā€œHuman brain gangliosides in development, agingand disease,ā€ International Journal of Developmental Biology,vol. 35, no. 3, pp. 289ā€“295, 1991.

    [159] R. B. Chan, T. G. Oliveira, E. P. Cortes et al., ā€œComparativelipidomic analysis of mouse and human brain with Alzheimerdisease,ā€ Journal of Biological Chemistry, vol. 287, no. 4, pp.2678ā€“2688, 2012.

    [160] C. G. Gottfries, I. Karlsson, and L. Svennerholm, ā€œMem-brane components separate early-onset Alzheimerā€™s diseasefrom senile dementia of the Alzheimer type,ā€ InternationalPsychogeriatrics, vol. 8, no. 3, pp. 365ā€“372, 1996.

    [161] M. Molander-Melin, K. Blennow, N. Bogdanovic, B. Dellhe-den, J. E. MaĢŠnsson, and P. Fredman, ā€œStructural membranealterations in Alzheimer brains found to be associated withregional disease development; increased density of gangliosidesGM1 and GM2 and loss of cholesterol in detergent-resistantmembrane domains,ā€ Journal of Neurochemistry, vol. 92, no. 1,pp. 171ā€“182, 2005.

    [162] L. Barrier, S. Ingrand, M. Damjanac, A. Rioux Bilan, J.Hugon, and G. Page, ā€œGenotype-related changes of gangliosidecomposition in brain regions of transgenic mouse models ofAlzheimerā€™s disease,ā€ Neurobiology of Aging, vol. 28, no. 12, pp.1863ā€“1872, 2007.

    [163] M. O. W. Grimm, E. G. Zinser, S. GroĢˆsgen et al., ā€œAmyloidprecursor protein (app) mediated regulation of gangliosidehomeostasis linking alzheimerā€™s disease pathology with gan-gliosidemetabolism,ā€ PLoSONE, vol. 7, no. 3, Article ID e34095,2012.

    [164] Q. Zha, Y. Ruan, T. Hartmann, K. Beyreuther, and D. Zhang,ā€œGM1 ganglioside regulates the proteolysis of amyloid precursorprotein,ā€Molecular Psychiatry, vol. 9, no. 10, pp. 946ā€“952, 2004.

  • BioMed Research International 15

    [165] O.Holmes, S. Paturi,W.Ye,M. S.Wolfe, andD. J. Selkoe, ā€œEffectsof membrane lipids on the activity and processivity of purifiedš›¾-secretase,ā€ Biochemistry, vol. 51, no. 17, pp. 3565ā€“3575, 2012.

    [166] I. Peters, U. Igbavboa, T. SchuĢˆtt et al., ā€œThe interaction of beta-amyloid protein with cellular membranes stimulates its ownproduction,ā€ Biochimica et Biophysica Acta, vol. 1788, no. 5, pp.964ā€“972, 2009.

    [167] I. Y. Tamboli, K. Prager, E. Barth, M. Heneka, K. Sandhoff, andJ. Walter, ā€œInhibition of glycosphingolipid biosynthesis reducessecretion of the š›½-amyloid precursor protein and amyloid š›½-peptide,ā€ Journal of Biological Chemistry, vol. 280, no. 30, pp.28110ā€“28117, 2005.

    [168] K. Yanagisawa, A. Odaka, N. Suzuki, and Y. Ihara, ā€œGM1ganglio