+ All Categories
Home > Documents > LevelsofSolubleApolipoproteinE/Amyloid- (A )Complex ... · gatesofA (oA ).Forthisstudy,anapoE/A...

LevelsofSolubleApolipoproteinE/Amyloid- (A )Complex ... · gatesofA (oA ).Forthisstudy,anapoE/A...

Date post: 02-Oct-2018
Category:
Upload: buimien
View: 214 times
Download: 0 times
Share this document with a friend
14
Levels of Soluble Apolipoprotein E/Amyloid- (A) Complex Are Reduced and Oligomeric A Increased with APOE4 and Alzheimer Disease in a Transgenic Mouse Model and Human Samples * S Received for publication, December 5, 2012, and in revised form, January 4, 2013 Published, JBC Papers in Press, January 4, 2013, DOI 10.1074/jbc.M112.442103 Leon M. Tai, a Tina Bilousova, b Lisa Jungbauer, a1 Stephen K. Roeske, a Katherine L. Youmans, a2 Chunjiang Yu, a Wayne W. Poon, c Lindsey B. Cornwell, c Carol A. Miller, d Harry V. Vinters, e3 Linda J. Van Eldik, f,g David W. Fardo, f,h Steve Estus, f,i Guojun Bu, j Karen Hoppens Gylys, b and Mary Jo LaDu a4 From the a Department of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois 60612, the b UCLA School of Nursing and Mary S. Easton Center for Alzheimer’s Research, Los Angeles, California 90095, the c Institute for Memory Impairments and Neurological Disorders, University of California at Irvine, Irvine, California 92697, the d Departments of Pathology, Neurology, and Program in Neuroscience, Keck University of Southern California School of Medicine, Los Angeles, California 90089, the e Department of Pathology and Laboratory Medicine, UCLA School of Medicine, Los Angeles, California 90095, the f Sanders-Brown Center on Aging, and the Departments of g Anatomy and Neurobiology, h Biostatistics, and i Physiology, University of Kentucky, Lexington, Kentucky 40536, and the j Department of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224 Background: An ELISA was developed to determine the role of apoE/A on soluble A accumulation. Results: In AD transgenic mouse brain and human synaptosomes and CSF, levels of soluble apoE/A are lower and oligomeric A levels are higher with APOE4 and AD. Conclusion: Isoform-specific apoE/A levels modulate soluble oligomeric A levels. Significance: ApoE/A and oligomeric A represent a mechanistic approach to AD biomarkers. Human apolipoprotein E (apoE) isoforms may differentially modulate amyloid- (A) levels. Evidence suggests physical interactions between apoE and A are partially responsible for these functional effects. However, the apoE/A complex is not a single static structure; rather, it is defined by detection methods. Thus, literature results are inconsistent and difficult to inter- pret. An ELISA was developed to measure soluble apoE/A in a single, quantitative method and was used to address the hypoth- esis that reduced levels of soluble apoE/A and an increase in soluble A, specifically oligomeric A (oA), are associated with APOE4 and AD. Previously, soluble A42 and oA levels were greater with APOE4 compared with APOE2/APOE3 in hip- pocampal homogenates from EFAD transgenic mice (express- ing five familial AD mutations and human apoE isoforms). In this study, soluble apoE/A levels were lower in E4FAD mice compared with E2FAD and E3FAD mice, thus providing evi- dence that apoE/A levels isoform-specifically modulate solu- ble oA clearance. Similar results were observed in soluble preparations of human cortical synaptosomes; apoE/A levels were lower in AD patients compared with controls and lower with APOE4 in the AD cohort. In human CSF, apoE/A levels were also lower in AD patients and with APOE4 in the AD cohort. Importantly, although total A42 levels decreased in AD patients compared with controls, oA levels increased and were greater with APOE4 in the AD cohort. Overall, apoE iso- form-specific formation of soluble apoE/A modulates oA lev- els, suggesting a basis for APOE4-induced AD risk and a mech- anistic approach to AD biomarkers. APOE4 is the primary genetic risk factor for Alzheimer dis- ease (AD), 5 although APOE2 reduces risk compared with APOE3. Although the mechanism(s) by which apolipoprotein E (apoE) and amyloid- peptide (A) affect the pathogenesis of AD remains unclear (1, 2), apoE isoform-specific physical inter- actions with A (apoE/A) may modulate the levels of A. These interactions appear to consist of two types, which may or may not be “on pathway” to amyloid deposition as follows: apoE isoform-specific effects on plaque development and apoE iso- form-specific effects on the levels of soluble, oligomeric aggre- * This work was supported, in whole or in part, by National Institutes of Health Grants P01AG030128 from NIA (to M. J. L. and S. E.), AG27465 (to K. H. G.), and G18879 (to C. A. M.). This work was also supported by Alzheimer’s Association Grant ZEN-08-899000 (to M. J. L.), University of Illinois at Chi- cago Center for Clinical and Translational Science Grant UL1RR029879 (to M. J. L.), and an Alzheimer’s Drug Discovery Foundation Grant (to M. J. L. and S. E.). This article was selected as a Paper of the Week. S This article contains supplemental Fig. S1 and Tables 1 and 2. 1 Present address: Medtronic, Inc., 7000 Central Ave. NE, RCE470, Minneapolis, MN 55432. 2 Present address: Dept. of Pharmacology, Boston University, Boston, MA 02218. 3 Supported by the Daljit S. and Elaine Sarkaria Chair in Diagnostic Medicine. 4 To whom correspondence should be addressed: University of Illinois at Chi- cago, Dept. of Anatomy and Cell Biology, 808 S. Wood St., M/C 512, Chi- cago, IL 60612. Tel.: 312-355-4795; Fax: 312-413-0354; E-mail: mladu@ uic.edu. 5 The abbreviations used are: AD, Alzheimer disease; A, amyloid-; apoE, apolipoprotein E; AUC, area under the curve; CSF, cerebrospinal fluid; FAD, familial AD; FAD-Tg, transgenic mice expressing FAD mutations; IHC, immunohistochemistry; 5FAD mice, FAD-Tg that co-express five FAD mutations; oA, oligomeric amyloid-; ROC; receiver operating character- istic curves; Tg, transgenic; T-Tau, total Tau; p-Tau-181, phosphorylated Tau at residue 181; ANOVA, analysis of variance; ISF, interstitial fluid. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 288, NO. 8, pp. 5914 –5926, February 22, 2013 © 2013 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. 5914 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013 by guest on October 1, 2018 http://www.jbc.org/ Downloaded from
Transcript

Levels of Soluble Apolipoprotein E/Amyloid-� (A�) ComplexAre Reduced and Oligomeric A� Increased with APOE4 andAlzheimer Disease in a Transgenic Mouse Model and HumanSamples*□S �

Received for publication, December 5, 2012, and in revised form, January 4, 2013 Published, JBC Papers in Press, January 4, 2013, DOI 10.1074/jbc.M112.442103

Leon M. Tai,a Tina Bilousova,b Lisa Jungbauer,a1 Stephen K. Roeske,a Katherine L. Youmans,a2 Chunjiang Yu,a

Wayne W. Poon,c Lindsey B. Cornwell,c Carol A. Miller,d Harry V. Vinters,e3 Linda J. Van Eldik,f,g David W. Fardo,f,h

Steve Estus,f,i Guojun Bu,j Karen Hoppens Gylys,b and Mary Jo LaDua4

From the aDepartment of Anatomy and Cell Biology, University of Illinois, Chicago, Illinois 60612, the bUCLA School of Nursing andMary S. Easton Center for Alzheimer’s Research, Los Angeles, California 90095, the cInstitute for Memory Impairments andNeurological Disorders, University of California at Irvine, Irvine, California 92697, the dDepartments of Pathology, Neurology, andProgram in Neuroscience, Keck University of Southern California School of Medicine, Los Angeles, California 90089, theeDepartment of Pathology and Laboratory Medicine, UCLA School of Medicine, Los Angeles, California 90095, the fSanders-BrownCenter on Aging, and the Departments of gAnatomy and Neurobiology, hBiostatistics, and iPhysiology, University of Kentucky,Lexington, Kentucky 40536, and the jDepartment of Neuroscience, Mayo Clinic, Jacksonville, Florida 32224

Background: An ELISA was developed to determine the role of apoE/A� on soluble A� accumulation.Results: In AD transgenic mouse brain and human synaptosomes and CSF, levels of soluble apoE/A� are lower and oligomericA� levels are higher with APOE4 and AD.Conclusion: Isoform-specific apoE/A� levels modulate soluble oligomeric A� levels.Significance: ApoE/A� and oligomeric A� represent a mechanistic approach to AD biomarkers.

Human apolipoprotein E (apoE) isoforms may differentiallymodulate amyloid-� (A�) levels. Evidence suggests physicalinteractions between apoE and A� are partially responsible forthese functional effects. However, the apoE/A� complex is not asingle static structure; rather, it is definedbydetectionmethods.Thus, literature results are inconsistent and difficult to inter-pret. An ELISA was developed to measure soluble apoE/A� in asingle, quantitativemethod andwas used to address the hypoth-esis that reduced levels of soluble apoE/A� and an increase insoluble A�, specifically oligomeric A� (oA�), are associatedwith APOE4 and AD. Previously, soluble A�42 and oA� levelswere greaterwithAPOE4 comparedwithAPOE2/APOE3 in hip-pocampal homogenates from EFAD transgenic mice (express-ing five familial AD mutations and human apoE isoforms). Inthis study, soluble apoE/A� levels were lower in E4FAD micecompared with E2FAD and E3FAD mice, thus providing evi-

dence that apoE/A� levels isoform-specifically modulate solu-ble oA� clearance. Similar results were observed in solublepreparations of human cortical synaptosomes; apoE/A� levelswere lower in AD patients compared with controls and lowerwith APOE4 in the AD cohort. In human CSF, apoE/A� levelswere also lower in AD patients and with APOE4 in the ADcohort. Importantly, although total A�42 levels decreased inAD patients compared with controls, oA� levels increased andwere greater with APOE4 in the AD cohort. Overall, apoE iso-form-specific formationof soluble apoE/A�modulates oA� lev-els, suggesting a basis for APOE4-induced AD risk and a mech-anistic approach to AD biomarkers.

APOE4 is the primary genetic risk factor for Alzheimer dis-ease (AD),5 although APOE2 reduces risk compared withAPOE3. Although themechanism(s) bywhich apolipoprotein E(apoE) and amyloid-� peptide (A�) affect the pathogenesis ofAD remains unclear (1, 2), apoE isoform-specific physical inter-actions with A� (apoE/A�) may modulate the levels of A�.These interactions appear to consist of two types, whichmay ormay not be “on pathway” to amyloid deposition as follows: apoEisoform-specific effects on plaque development and apoE iso-form-specific effects on the levels of soluble, oligomeric aggre-

* This work was supported, in whole or in part, by National Institutes of HealthGrants P01AG030128 from NIA (to M. J. L. and S. E.), AG27465 (to K. H. G.),and G18879 (to C. A. M.). This work was also supported by Alzheimer’sAssociation Grant ZEN-08-899000 (to M. J. L.), University of Illinois at Chi-cago Center for Clinical and Translational Science Grant UL1RR029879 (toM. J. L.), and an Alzheimer’s Drug Discovery Foundation Grant (to M. J. L.and S. E.).

� This article was selected as a Paper of the Week.□S This article contains supplemental Fig. S1 and Tables 1 and 2.1 Present address: Medtronic, Inc., 7000 Central Ave. NE, RCE470, Minneapolis,

MN 55432.2 Present address: Dept. of Pharmacology, Boston University, Boston, MA

02218.3 Supported by the Daljit S. and Elaine Sarkaria Chair in Diagnostic Medicine.4 To whom correspondence should be addressed: University of Illinois at Chi-

cago, Dept. of Anatomy and Cell Biology, 808 S. Wood St., M/C 512, Chi-cago, IL 60612. Tel.: 312-355-4795; Fax: 312-413-0354; E-mail: [email protected].

5 The abbreviations used are: AD, Alzheimer disease; A�, amyloid-�; apoE,apolipoprotein E; AUC, area under the curve; CSF, cerebrospinal fluid; FAD,familial AD; FAD-Tg, transgenic mice expressing FAD mutations; IHC,immunohistochemistry; 5�FAD mice, FAD-Tg that co-express five FADmutations; oA�, oligomeric amyloid-�; ROC; receiver operating character-istic curves; Tg, transgenic; T-Tau, total Tau; p-Tau-181, phosphorylatedTau at residue 181; ANOVA, analysis of variance; ISF, interstitial fluid.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 288, NO. 8, pp. 5914 –5926, February 22, 2013© 2013 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A.

5914 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

gates of A� (oA�). For this study, an apoE/A�ELISAwas devel-oped to determine the effect of theAPOE genotype on the levelsof soluble apoE/A� and A�.The amyloid hypothesis posits that deposition of extracellu-

lar amyloid is central for producing the neurodegenerative pro-cesses characteristic of AD (3). In the landmark 1992 paper,Wisniewski and Frangione (4) proposed that apoE was a “path-ological chaperone,” based on the co-localization of apoE withA� in amyloid plaques as detected via immunohistochemistry(IHC). Thus, apoE was thought to facilitate the process of A�deposition as amyloid. Biochemical analyses validate IHCmeasures, as the levels of apoE and A� are equivalent in theinsoluble extraction fraction from brains of transgenic miceexpressing familial AD (FAD)mutations (FAD-Tg), specificallythe 5�FAD-Tg mice (5). The association of APOE4 with ADrisk was first described in 1993 (6, 7), leading to research effortsfocused on the effects of the APOE genotype on plaque burdenand the structural relationship between apoE and amyloid. IHCanalysis demonstrates that plaque deposition is greater withAPOE4 compared with APOE3 in AD and nondemented con-trols (8, 9) and that a higher proportion of A�within a plaque isassociatedwith apoE4 thanwith apoE3 (10). Biochemical anal-ysis confirms that the levels of apoE and A� are also higherwith APOE4 compared with APOE3 in the insoluble extrac-tion fraction from brains of FAD-Tgmice (11). Thus, APOE4not only facilitates amyloid deposition but also forms agreater amount and/or more stable form of apoE4/amyloidthan apoE3/amyloid.The amyloid hypothesis has been revised, as plaque burden

does not correlate with the dementia that is characteristic ofAD (12, 13). However, soluble A� and oA� do correlate withcognitive decline and disease severity in humans (14). oA� isalso detected in FAD-Tg mice and is associated with memorydecline (14). Thus, the structure-function relationship of solu-ble A� and oA� is an area of intense research. However, unlikeamyloid, which refers to a specific parallel �-sheet structure,oA� refers to a number of assemblies defined by a variety ofdetection methods (14). This makes interpretation and com-parison of results problematic, particularly with in vivo data.We recently developed anoA�ELISAanddemonstrated that inEFAD-Tg mice soluble A�42 and oA� are greater in E4FADmice, compared with E2FAD and E3FAD (15). A� clearancealso appears to be decreased with APOE4 (16), suggesting thatsoluble apoE/A� may modulate soluble A� and oA� levels.

Research efforts to determine apoE/A� levels, particularlysoluble complex levels, have been hindered by a lack of quanti-tative detection methods. A variety of techniques have pro-duced results that can be inconsistent and difficult to interpret(7, 17–27). Even the initial biochemical characterizations of themolecular interactions between apoE and A� were problem-atic, primarily because of two parameters. The first variable wasthe lipidation state of apoE. Using purified protein, apoE4bound A� with a higher affinity than apoE3 (28, 29). However,this result is reversed using physiologically relevant, lipidatedapoE; levels of the apoE3/A� complex are significantly greaterthan the apoE4/A� complex (21, 28, 29). Second, the definitionof an apoE/A� is primarily operational, with assay stringencythe primary variable (7, 17–27, 30). For example, apoE3/A�

levels are greater than apoE4/A� as determined by Westernanalysis of SDS-PAGE (21), but by nondenaturing gel electro-phoresis, the levels of the apoE3/A� complex are comparablewith apoE4/A� (31). Although these data are consistent with anSDS-stable apoE3/A� complex (32), and an apoE4/A� complexthat is disrupted by SDS, the total amount of apoE/A� cannotbe quantified byWestern analysis of SDS-PAGE. The first goalof this study was to define biochemically generated apoE/A� inthe context of a single quantitative and potentially highthroughputmethod thatwould also define both total and deter-gent (SDS)-stable apoE/A�, providing a platform for compari-son among apoE isoforms and across methods. Thus, a newapoE/A� ELISA was developed and optimized biochemically.In vitro, total complex levels were equivalent among the apoEisoforms, although the apoE3/A� complex was more SDS-sta-ble than the apoE4/A� complex but was less SDS-stable thanapoE2/A�. These results are consistent with previous resultsutilizing several methods that suggest the levels of apoE3/A�and apoE4/A� complex are comparable in the absence of SDSbut that SDS-stable apoE3/A� complex levels are greater thanapoE4/A� (18, 21, 22, 27).In contrast to biochemical analysis, the number of in vivo

reports on soluble apoE/A� is limited. ApoE/A� complex hasbeen detected in the soluble fraction of human brain (33) and inhuman cerebrospinal fluid (CSF) (30, 34), although the datawere primarily produced usingWestern analysis of SDS-PAGE.By IHC, apoE also co-localizes with A� at the synapse (35), andinsoluble apoE/A� complexes appear to form preferentiallywith apoE4 compared with apoE3 (36). However, the effect oftheAPOE genotype on soluble synaptic apoE/A� levels remainsunclear (37–40). Thus, the new apoE/A� ELISA was used invivo to determine the levels of soluble apoE/A� and the effect ofthe APOE genotype. In EFAD transgenic mice, previous datademonstrated that with APOE4 the soluble A�42 and oA� lev-els were greater (15), and in the data presented herein, solubleapoE4/A� complex levels were lower and less stable comparedwith apoE3/A� and apoE2/A� levels. In human synaptosomepreparations and CSF, apoE/A� levels were lower in AD com-pared with controls and withAPOE4 compared withAPOE3 inthe AD cohort. Importantly, in human CSF, although totalA�42 levels decreased in AD patients compared with controls,oA� levels increased and were greater with APOE4 in the ADcohort. Taken together, the low levels of the soluble apoE4/A�complex and high levels of the soluble oA� suggest an impairedclearance mechanism for the soluble forms of A� and a poten-tial basis for APOE4-induced AD risk, as well as a mechanisticapproach to CSF biomarkers for AD.

EXPERIMENTAL PROCEDURES

Materials

High bind plates (MaxisorpTM) and low bind plates(MicrowellTM) were purchased from NUNC, Rochester, NY.Anti (�)-A� antibodies used were as follows: 6E10 (CovanceLabs, Madison, WI); 4G8 (Senetek, Maryland Height, MD),and MOAB-2 (41) (LaDu Laboratory and available fromAbcam, Cambridge, MA; Biosensis, Temecula, CA; Millipore,Bilerica, MA; Novus, Littleton, CO; and Cayman, Ann Arbor

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5915

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

MI). Goat �-apoE antibodies were from Calbiochem, Meridian(Memphis, TN), and Millipore (Billerica, MA). RecombinantapoE3 was from BioVision (Milpitas, CA), and synthetic A�peptides were from California Peptide (Napa, CA).

HEK-ApoE

ApoE from HEK-293 cells stably transfected with humancDNA encoding apoE2, apoE3, or apoE4 was prepared asdescribed previously (21, 42, 43). Briefly, serum-free condi-tioned media were concentrated �50-fold (Centriprep, Ami-con, Inc.) and fractionated by size exclusion chromatography.The resulting fractions containing apoE particles were pooledand the concentration of apoE quantified.

Amyloid-� (A�) Peptide

A� peptides were prepared as described previously (44–46).1,1,1,3,3,3-Hexafluoro-2-propanol-treated A� was dissolved inDMSO to 5 mM and then to 100 �M in phenol red-free F-12media (BioSource, Camarillo, CA) for unaggregated and oA� or10 mM HCl for fibrillar A�. Unaggregated A� was freshly pre-pared just prior to use; oA�42 preparations were aged for 24 hat 4 °C and fibrillar A�42 preparations for 24 h at 37 °C.

ApoE/A� Complex Standard Development and BiochemicalCharacterization

ApoE/A� Complex Formation—HEK-apoE or recombinantapoE and A� were incubated at the indicated concentrationsfor 2 h at room temperature, pH 7.4, with SDS (Sigma) or vehi-cle at the indicated concentrations. The pHprofile for apoE/A�levels was conducted as described (21).ELISA Curve Fitting—In the absence of SDS, the EC50 value

for A� and apoEwas calculated using the four-parameter logis-tic Equation 1,

apoE/A� complex levels � bottom � �top � bottom�/

�1 � 10∧��logEC50 � X� � Hill slope�� (Eq. 1)

Top and bottom represents the apoE/A� levels at the plateaus.EC50 is the concentration of A� or apoE that produces 50%maximal response.X is the concentration of the variable i.e.A�or apoE.In the presence of SDS, the IC50 value for SDS was calculated

according to Equation 2,

apoE/A� complex levels �% of control�

� 100/�1 � 10∧�X � logIC50�� (Eq. 2)

IC50 is the effective concentration of SDS that produces 50%response. X corresponds to the concentration of SDS.Analysis was conducted for each individual experiment, and

data were analyzed using one-way analysis of variance(ANOVA) followed by Tukey’s post hoc analysis GraphPadPrism Version 5 for Macintosh was used for all curve-fittinganalyses.

ApoE/A� Complex ELISA

Biochemical ELISA Development—To accurately quantifytotal and SDS-stable levels of apoE/A�, a specific ELISA was

developed. The apoE/A� complex formed between HEK-apoEand unaggregated A�42 was utilized for ELISA development.To minimize nonspecific binding of apoE and A� and to max-imize apoE/A� detection, a number of antibody combinationswere screened as capture or detection antibodies on NUNCMaxisorpTM (high bind) or MicrowellTM (low bind) plates(supplemental Fig. 1). Results demonstrated the following. 1)Nonspecific binding of A� to high and low bind plates pre-cludes the use of �-A� antibodies for detection. 2) Nonspecificbinding of HEK-apoE prevents the use of high bind plates (sup-plemental Fig. 1A). 3) ApoE/A� complex, but not apoE orA�, isdetected on low bind plates using �-apoE capture and �-A�detection antibodies (supplemental Fig. 1B). 4) �-A�(MOAB-2) capture and �-apoE (Calbiochem) detection anti-bodies produce the highest signal/background ratio comparedwith other antibodies tested (supplemental Fig. 1, C and D).Thus, the optimal reagents/conditions for specific HEK-apoE/A� detection by ELISA were low bind 96-well plates with�-A� (MOAB-2) capture and �-apoE (Calbiochem) detectionantibodies.ApoE/A� ELISA—For protocol 1, low bind plates were

coated withMOAB-2 at 6.25�g/ml in carbonate coating bufferovernight at 4 °C. Plates were washed (three times in PBS),blocked (4% BSA, 1.5 h, 37 °C), washed again (three times inPBS), and incubated with samples overnight. The plates werethen washed (three times in PBS), incubated with a 200-folddilution of �-apoE (Calbiochem) (1.5 h, 37 °C), washed, andincubated with HRP-conjugated antibodies (1.5 h, RT, 1:5000dilution, Jackson ImmunoResearch, West Grove, PA). Follow-ing a final wash step (three times in PBS), 3,3�,5,5�-tetrameth-ylbenzidine liquid substrate Superslow (Sigma) was added, andabsorbance wasmeasured atA620. For protocol 2, all steps wereidentical to protocol 1, with the exception that high bind plateswere utilized (see ELISA analysis of human CSF).

Soluble ApoE/A� Complex Detection in EFAD Mice

EFAD Transgenic Mice—Experiments follow the UIC Insti-tutional Animal Care and Use Committee protocols. EFADmice (15) are the result of crossing 5�FADmice, which co-ex-press five FAD mutations (APP K670N/M671L, I716V, andV717I and PS1, M146L and L286V) under the control of theThy-1 promoter with apoE-targeted replacement mice. Detailson the production, breeding, genotyping, and genetic back-ground of these mice are described in Ref. 15.Tissue Preparation—Brain tissue isolation and serial protein

extraction were conducted as described previously (5, 15).Briefly, 6-month-old male EFAD mice were anesthetized withsodium pentobarbital (50 mg/kg) and transcardially perfused(PBS plus protease inhibitors (Calbiochem, set 3)), and brainswere removed and dissected at the midline. Right hemi-brainswere dissected on ice into cortex, hippocampus, and cerebel-lum, immediately snap-frozen in liquid nitrogen, and stored at�80 °C until use. The dissected tissue was homogenized in 15volumes (w/v) of TBS; samples were centrifuged (100,000 � g,1 h at 4 °C), and the TBS (soluble) fraction was aliquoted priorto freezing in liquid nitrogen and storage at �80 °C.ApoE/A� Complex—The apoE/A� levels were measured

using a 4-fold sample dilution of the TBS extraction fraction

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5916 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

from the hippocampus of EFAD mice according to apoE/A�ELISA protocol 1. The standard curve used a fixed HEK-apoEconcentration of 140 nM (apoE concentration in the TBSextraction of EFAD mice at 6 months for all APOE genotypes)and variedA� concentrations. Datawere normalized to proteinconcentration in each sample.

ApoE/A� Detection in Human Synaptosomes

Brain samples of parietal cortex (A7, A39, and A40) wereobtained at autopsy for cases followed by the Alzheimer diseaseresearch centers at UCLA, University of California at Irvine,and University of Southern California (supplemental Table 1);the last clinical diagnosis and full neuropathological report anddiagnosis were available for all cases. Control samples includednormal cases and pathological controls. Immediately uponreceipt, samples (�0.3–5 g) wereminced in 0.32 M sucrose withprotease inhibitors (2mM EDTA, 2mM EGTA, 0.2mM PMSF, 1mM sodium pyrophosphate, 5 mM NaF, 10 mM Tris) and thenstored at �70 °C until homogenization. The P-2 (crude synap-tosome; synaptosome-enriched fraction) was prepared asdescribed previously (42); briefly, tissue was homogenized inice-cold buffer (0.32 M sucrose, 10 mM Tris, pH 7.5, plus prote-ase inhibitors: pepstatin (4mg/ml), aprotinin (5mg/ml), trypsininhibitor (20 mg/ml), EDTA (2 mM), EGTA (2 mM), PMSF (0.2mM), leupeptin (4 mg/ml)). The homogenate was first centri-fuged at 1000 � g for 10 min; the resulting supernatant wascentrifuged at 10,000 � g for 20 min to obtain the crude synap-tosomal pellet. Aliquots of P-2 were routinely cryopreserved in0.32 M sucrose and banked at�70 °C until the day of the exper-iment. On the day of the experiment, cryopreserved human P-2aliquots were defrosted at 37 °C, resuspended in PBS with pro-tease inhibitors, sonicated, and centrifuged for 4 min at 6000rpm. Supernatant was collected, and total protein concentra-tion was defined using BCA protein assay (Pierce). ApoE/A�complex levels were measured using a 5-fold sample dilutionaccording to apoE/A� ELISA protocol 1. The standard curveused a fixed HEK-apoE concentration of 14 nM (apoE concen-tration in the synaptosomes) and varied A� concentrations.Human data were normalized according to total protein con-centration in each sample.

ELISA Analysis of Human CSF

CSF samples were obtained at autopsy at the Alzheimer Dis-ease Center at the University of Kentucky (supplemental Table2). Diagnoses of AD and non-AD were performed at a consen-sus conference of the AD Center Neuropathology and ClinicalCores and were based upon evaluation of both cognitive status,i.e. Clinical Dementia rating and Mini-Mental State Examina-tion (MMSE) scores, as well as neuropathology, i.e.Braak stagesthat rate the extent of neurofibrillary pathology into the neo-cortex and the NIAReagan Institute neuropathology classifica-tion, which includes counts of both neuritic senile plaques andneurofibrillary tangles and provides a likelihood staging of ADneuropathological diagnosis (47, 48). For ELISA analysis, A�42,total Tau (T-Tau), and phosphorylated Tau 181 (p-Tau-181)levels weremeasured using Innotest�ELISA kits (Innogenetics,Gent, Belgium) according to themanufacturer’s protocol; apoElevels were measured using �-apoE (Millipore) as capture and

�-apoE (Meridian) as detection as described (11). oA� levelswere measured using MOAB-2 capture (5) and biotinylatedMOAB-2 as detection antibody as described previously (15).ApoE/A� complex levels were measured using a 2-fold sampledilution according to apoE/A� ELISA protocol 2, with a stand-ard curve of 5 �g/ml recombinant apoE (reported CSF apoEconcentration) and varied A� concentrations.

Statistical Analysis

Datawere analyzed by one-wayANOVA followed byTukey’spost hoc analysis (Figs. 2, A and B, and 3–5) or by two-wayANOVA followed by Bonferroni post hoc analysis (Fig. 2C).Correlation analysis was conducted using Spearman’s correla-tion (Fig. 5, E and F). All data were analyzed using GraphPadPrism version 5 for Macintosh, and p � 0.05 was consideredsignificant. Receiver operating characteristic (ROC) curves(Fig. 6)were constructed for eachmarker using the pROCpack-age in R (49, 50). Areas under the curves were compared by themethod of DeLong et al. (51).

RESULTS

Biochemical Development of ApoE/A� Complex ELISA—Ini-tially, biochemical analysis using HEK-apoE and synthetic A�preparations (Fig. 1) (45) was conducted to validate theapoE/A� ELISA and to determine the effect of apoE isoform onsoluble apoE/A� levels and stability.Total ApoE/A� Complex Levels Are Not Affected by ApoE

Isoform—Total complex levels were measured in samples con-taining a fixed apoE concentration (30 nM) and a varied concen-tration of unaggregated A�42 (0.15–150 nM) (Fig. 1A) or usingvaried apoE concentration (0–1500 nM) and a fixedA� concen-tration (3 nM) (Fig. 1B). Overall, apoE/A� levels were saturableand dependent on apoE and A� concentrations but not apoEisoform. Indeed, when these data were analyzed using a four-parameter logistic equation, which is appropriate for analyzingELISA saturation curves (52), there were no differencesbetween the calculated EC50 values among the apoE isoforms(�3 nM for A� in Fig. 3A and�30 nM for apoE in Fig. 1B). TotalapoE/A� levels were also equivalent for apoE2, apoE3, andapoE4 with unaggregated A�40, oA�42, and fibrillar A�42(data not shown). Thus, apoE isoform does not determine totalapoE/A� levels biochemically.ApoE2/A� and ApoE3/A� Complex Exhibits Greater Stabil-

ity than ApoE4/A� Complex—As apoE isoform did not affecttotal complex levels when assessed by ELISA, SDSwas added tosamples as a measure of stability (Fig. 1C). ApoE and A� wereincubated for 2 h at concentrations that correspond to the EC50values identified for total apoE/A� levels, specifically 3 nMA�42 and 30 nM apoE, and then SDS was added over a range ofconcentrations (up to 2%). Complex stability from highest tolowest was apoE2/A� � apoE3/A� � apoE4/A�. This was evi-dent as the SDS IC50 value was 1.5-fold higher for apoE2/A�complex and 3-fold lower for the apoE4/A� complex comparedwith the apoE3/A� complex. In addition to SDS, the apoE4/A�complex was less stable at mildly acidic pH (5) than apoE2/A�and apoE3/A� complex (Fig. 1D). Therefore, apoE/A� levelswere not determined by the apoE isoform; however, the

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5917

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

apoE4/A� complex is less stable, and the apoE2/A� is morestable than the apoE3/A� complex.Soluble ApoE/A� Complex Levels in EFAD Mice—To deter-

mine the effect of APOE genotype on soluble apoE/A� levels,the tractable EFAD mouse model was utilized. For this study,apoE/A� levels were measured in the soluble hippocampalhomogenates from EFAD mice at 6 months (Fig. 2), an agewhere soluble oA� levels are greater in E4FAD (APOE4) com-pared with E2FAD (APOE2) and E3FAD (APOE3) mice (15).

ApoE/A� Complex ELISA Optimization in EFADMice—Ini-tially, soluble apoE/A� detection by ELISA was validated usingE3FADmice at 6 months (Fig. 2A). For a quantitative standardto enable cross-plate comparisons, the complex formedbetween HEK-apoE3 at a fixed concentration of 140 nM, whichcorresponds to soluble apoE levels in EFAD mice at 6 months,and a varied concentration of unaggregated A�42 was utilized.Specific soluble apoE/A� levels were detected by ELISA as fol-lows. 1) Soluble hippocampal apoE/A�was only detected using

FIGURE 1. Biochemical characterization of apoE/A� ELISA with HEK-apoE and synthetic A�. A, total apoE/A� levels for each apoE isoform with HEK-apoEfixed at 30.0 nM and unaggregated A�42 varied from 0.15 to 150.0 nM. B, total level of apoE/A� for each apoE isoform with unaggregated A�42 fixed at 3.0 nM

and HEK-apoE varied from 1.5 to 1500.0 nM. C, stability of apoE/A� in the presence of SDS from 0 –2%. D, stability of apoE/A� at varied pH. For all experimentsn 5 with duplicate samples. Data are expressed as mean S.E., analyzed by one-way ANOVA with Tukey’s multiple comparison post hoc analysis. *, p � 0.05compared with apoE3;#, p � 0.05 compared with apoE2.

FIGURE 2. ApoE/A� complex levels and stability in soluble brain extracts from EFAD mice. A, standardization and control for apoE/A� levels in the soluble (TBS)extraction fraction from the hippocampus and cerebellum of E3FAD mice at 6 months. B, apoE/A� complex in the soluble extraction fraction from the hippocampusof E2FAD, E3FAD, and E4FAD mice at 6 months. C, soluble apoE/A� stability in 0.02% and 0.2% SDS in samples as described for B. Standard curve for apoE/A� ELISA,140.0 nM HEK-apoE3 with 0.15–50.0 nM unaggregated A�42. For all experiments n 8 with duplicate samples. Data are expressed as mean S.E. and were analyzedby one-way ANOVA followed by Tukey’s post hoc analysis (B), or by two-way ANOVA followed by Bonferroni post hoc analysis (C). *, p � 0.05.

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5918 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

MOAB-2 as a capture antibody, as no signal was seen whenusing a nonspecific IgG2b isotype-matched capture antibody. 2)Complex levels decreased with increased sample dilution. 3)There were no detectable soluble complex levels in the cerebel-lum, a region spared of A� pathology in EFADmice. These datavalidate soluble apoE/A� detection in vivo by ELISA.Soluble ApoE/A� Complex Levels Are Lower and Less Stable

with APOE4—Next, the effect of APOE genotype on solubleapoE/A� levels and stability was determined. ApoE/A� com-plex levels were 50% lower in E4FAD mice compared withE2FAD and E3FAD mice (Fig. 2B). For apoE/A� stability (Fig.2C), complex levelsweremeasured from the same sample in thepresence of 0, 0.02, or 0.2% SDS. Complex levels were normal-ized to the 0% SDS control for each paired samples set. Theaddition of SDS reduced apoE/A� levels, in anAPOE genotype-specific manner. With 0.02% SDS, apoE4/A� complex levelswere reduced by �60%, apoE3/A� complex levels by �50%,and apoE2/A� complex levels by �30%. The addition of 0.2%SDS further lowered complex levels in E3FADandE4FADmicebut not E2FADmice. These data demonstrate that soluble hip-pocampal apoE/A� levels are lower and less SDS-stable inE4FADmice compared with E3FAD and E2FADmice and thatthe apoE2/A� complex is more stable than the apoE3/A�complex.Soluble ApoE/A� Complex in Synaptosomes—To determine

the effect of AD and APOE genotype on soluble synapticapoE/A� levels, cortical synaptosomes were isolated from con-trol (APOE3/3 and APOE4/X) and AD patients (APOE3/3 andAPOE4/X) (Fig. 3).ApoE/A� Complex Levels Were Lower in AD Patients Com-

pared with Controls and with APOE4 in the AD Cohort—In theabsence of SDS (Fig. 3A), the data are normalized to APOE3/3controls. In the control individuals, there was no significantdifference between apoE/A� levels in the APOE3/3 andAPOE4/X. In the AD patients, apoE/A� levels were signifi-cantly lower, i.e. 70% lower forAPOE3/3ADpatients compared

with APOE3/3 controls and 90% lower in APOE4/X ADpatients compared with APOE4/X controls (Fig. 3A). In addi-tion, within the AD cohort, total apoE/A� levels were 66%lower with APOE4/X compared with APOE3/3.

To address the SDS stability of the apoE/A� (Fig. 3B), com-plex levels were measured from the same sample in the pres-ence of 0 or 0.02% SDS. ApoE/A� complex levels were thennormalized to the 0% SDS control for each paired sample set. Inthe control individuals, the addition of SDS results in a signifi-cant decrease in apoE/A� levels withAPOE4/X compared withAPOE3/3. ApoE/A� complex stability was not significantly dif-ferent in the APOE4/X AD patients compared with APOE3/3AD patients. This is primarily due to the very low levels ofcomplex present in APOE4/X AD samples in the absence ofSDS (Fig. 3A); thus, after the addition of SDS, any further reduc-tion results in values for the complex that are at the limit ofdetection for this ELISA. Again, this results from the pairwisecomparison between apoE/A� levels in APOE4/X AD patientsin the absence of SDS (for example Fig. 3A), where the levels ofapoE/A� are already low, and apoE/A� levels in the presence ofSDS (Fig. 3B).ApoE/A� Complex in Human CSF—The CSF is as an indica-

tion of the concentration of soluble proteins in the brain paren-chyma. Therefore, the hypothesis that reduced levels of solubleapoE/A� and an increase in soluble oA� levels are associatedwithAD and APOE4 was tested in post-mortem CSF samples fromcontrol (APOE3/3) and AD patients (APOE3/3 andAPOE4/4).Oligomeric A� Levels Were Higher in AD Patients Compared

with Controls and Significantly Greater with APOE4 within theADCohort—Recently, we described an oA� ELISA that detectsoA� levels in EFADmice (15), using a previously described oA�preparation (53). For this study, a protocol characterized by ourlaboratory (46) was used to produce the oA� standard and isshown compared with fibrillar and unaggregated A�42 prepa-rations (Fig. 4A). As demonstrated in Fig. 4A, the oA� ELISAdemonstrates concentration-dependent detection of oA�, with

FIGURE 3. Soluble apoE/A� levels and stability in synaptosome-enriched extracts from human cortex. A and B were measured in age-matched controlsubjects (APOE3/3 and APOE4/X) and Alzheimer disease (AD) patients (APOE3/3 and APOE4/X). Description of sample groups is shown in supplemental Table 1.A, apoE/A� complex levels in cortical synaptosomes (P-2 fraction). Controls, n 10 APOE3/3, n 7 APOE4/X; AD, n 9 APOE3/3, n 7 APOE4/X. B, apoE/A�complex stability in 0.02% SDS. Standard curve for apoE/A� ELISA in human synaptosome-enriched extracts: 14.0 nM HEK-apoE3 with 0.86 – 450.0 nM unag-gregated A�42. Controls, n 5 APOE3/3, n 5 APOE4/X; AD, n 6 APOE3/3, n 5 APOE4/X. Data are expressed as mean S.E., analyzed by one-way ANOVAfollowed by Tukey’s multiple comparison post hoc analysis. *, p � 0.05.

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5919

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

a significantly lower affinity for fibrillar A�42 and no detectionof unaggregated A�42.This ELISA was used to determine whether oA� levels were

influenced byADdiagnosis orAPOE genotype. oA� levels weresignificantly increased in AD patients compared with controls,and importantly, oA� levels were significantly greater inAPOE4/4 AD patients compared with APOE3/3 AD patients(Fig. 4B). For comparative purposes, the established AD bio-markers A�42 (Fig. 4C), total Tau (T-Tau) (Fig. 4D), and phos-phorylated Tau 181 (p-Tau-181) (Fig. 4E) levels weremeasuredby ELISAs (54) in the same samples as oA�. As expected, A�42levels were significantly lower, and p-Tau-181 levels signifi-cantly greater in both the AD groups (APOE3/3 and APOE4/4)compared with age-matched controls (APOE3/3). T-Tau levelswere significantly greater in the APOE4/4 AD patients but notthe APOE3/3 AD patients compared with the APOE3/3 con-trols. Of particular interest, in the AD group, APOE genotypedid not affect the levels ofA�42, T-Tau, or p-Tau-181, all estab-lished AD biomarkers. Thus, oA� levels may play a role in ADprogression, in an APOE genotype-specific manner.ApoE/A� Complex ELISA Optimization for Human CSF—

An important consideration for measuring specific proteins inthe CSF by ELISA is a standard to allow quantification of sam-ples used on different microplates, across studies, so as to allowfuture retrospective analysis. Although HEK-apoE is an impor-tant apoE source for biochemical studies, the relatively longsample preparation time, potential intra-laboratory differencesin production quality, long term stability issues, and lack ofcommercialization hinders routine use as an apoE/A� stan-dard. Therefore, apoE/A� formed between recombinant

apoE3, at concentrations corresponding to those in humanCSF(5 �g/ml), and varied unaggregated A�42 concentrations wasused as a standard curve for the CSF samples (Fig. 5A). SpecificapoE/A� detection by ELISA in humanCSF (APOE3/3 control)was demonstrated by a high signal with capture antibody(MOAB-2) that decreased proportionately to sample dilutionand no observed signal without a capture antibody, all usinghigh bind plates to increase sensitivity (Fig. 5B).ApoE/A� Complex Levels Were Lower in CSF from AD

Patients Compared With Controls and, Importantly, Signifi-cantly Lower with APOE4 within the AD Cohort—ApoE/A�complex levels may modulate soluble oA� levels in the CNS.The increased oA� levels by AD and APOE4 raised the impor-tant question of what was the effect on apoE/A� levels.ApoE/A� complex levels were significantly lower in both ADcohorts compared with the control group (Fig. 5C). Impor-tantly, complex levels were significantly lower in APOE4/4 ADpatients compared with the APOE3/3 AD patients. Thus,APOE4 did affect the levels of oA� (increased) and apoE/A�(decreased) in the AD cohort and did not affect the levels ofA�42, T-Tau, or p-Tau-181, suggesting oA� and apoE/A� lev-els may play a role in AD progression. Although apoE levelswere lower in AD patients compared with controls (Fig. 5D),there was no correlation between apoE/A� levels and eitherapoE (Fig. 5E, Spearman’s r value 0.27, p 0.25) or A�42 (Fig.5F, Spearman’s r value 0.04, p 0.87) levels in the AD patientsample set. This suggests that the levels of apoE/A� are inde-pendent of the values of its two components. Thus, apoE/A�levels are affected by both AD and APOE genotype.

FIGURE 4. Oligomeric A� in human CSF compared with A�42, T-Tau, and p-Tau-181. A, standard curve for oligomeric A� ELISA: 0 –500 ng/ml oligomeric,fibrillar, and unaggregated A�42 preparations. B–E were measured in the CSF from age-matched control subjects (APOE3/3) and AD patients (APOE3/3 andAPOE4/4). Description of sample groups is shown in supplemental Table 2. B, oA�. C, A�42. D, T-Tau; E, p-Tau-181. Innotest� kits by Innogenetics for A�42,T-Tau, and p-Tau-181. For all experiments n 10 with duplicate samples. Data are expressed as mean S.E., analyzed by one-way ANOVA followed by Tukey’smultiple comparison post hoc analysis. *, p � 0.05. L.O.D. limit of detection.

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5920 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

oA� and ApoE/A� Complex as AD Biomarkers—In addi-tion to a potential mechanistic interpretation for AD pro-gression, oA� and apoE/A� levels may act as AD biomarkers.As APOE genotype affects oA� and apoE/A� levels, the opti-mal method for assessing the diagnostic potential of thesemarkers is analysis in control and AD patients with theAPOE3/3 genotype. ROC curves were utilized to determinethe predictive accuracy of each marker (Fig. 6). The ROCcurves are constructed by varying the threshold to classifypredicted AD cases and controls. Predicted probabilities ofbeing an AD case are calculated from marginal logisticregression models, and sensitivity (the proportion of ADcases correctly predicted, y axis) and specificity (the propor-tion of AD controls correctly predicted, i.e. true negativerate, x axis) are calculated based on each subject’s predictedcase probability being above or below the varying threshold,respectively. The area under the curve (AUC) of the ROC

curves is calculated, and an AUC of 0.5 demonstrates noinformation or diagnostic ability, whereas the higher theAUC is above 0.5, the greater the diagnostic accuracy of thebiomarker. ROC analysis demonstrated the potential use ofboth oA� and apoE/A� as AD biomarkers, with AUCs of 0.7and 0.875, respectively. Next, the ROC AUCs of oA� andapoE/A� were compared with the traditional AD biomarkersas follows: A�42, p-Tau-181, and T-Tau. A�42 was signifi-cantly more predictive of AD (p � 0.05) compared with theother markers except apoE/A� (p 0.14). Both oA� (p 0.70) and apoE/A� (p 0.41) complexes were as predictivefor AD as p-Tau-181, and apoE/A� was more predictive thanT-Tau (p � 0.012). Overall, in control and AD patients withthe APOE3/3 genotype, the estimated predictive ability forAD based on the AUC values for each marker was A�42(AUC 0.98) � apoE/A� (0.875) � p-Tau-181 (0.775) �oA� (0.7) � T-Tau (0.59).

FIGURE 5. ApoE/A� complex levels in human CSF. A, standard curve for apoE/A� ELISA in human CSF, 5 �g/ml recombinant apoE3 with 0.15–50.0 ng/mlunaggregated A�42. B, specific apoE/A� detection by ELISA in control human APOE3/3 CSF. The standards and CSF samples were diluted 2–16-fold, andapoE/A� levels were measured. C, apoE/A� complex; D, apoE levels were measured in age-matched control subjects (APOE3/3) and AD patients (APOE3/3 andAPOE4/4). Description of sample groups is shown in supplemental Table 2. For all experiments, n 10 with duplicate samples. Data are expressed as mean S.E., analyzed by one-way ANOVA followed by Tukey’s multiple comparison post hoc analysis. *, p � 0.05. L.O.D. limit of detection. Spearman’s correlationanalysis between apoE/A� and apoE (E) or A�42 (F) in CSF of AD patients (APOE3/3 and APOE4/4).

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5921

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

DISCUSSION

For this study, a quantitative apoE/A� ELISA was developedand characterized biochemically and then applied in vivo todetermine the effect of the APOE genotype and AD on solublelevels of apoE/A� complex and oA�. Soluble levels of oA� arehigher, and apoE/A� are lower with AD and specificallyAPOE4.Biochemical data using HEK-apoE demonstrate that the

apoE isoformdoes not affect total levels of apoE/A� but that theapoE4/A� complex is less stable than the apoE3/A� complex.By measuring total and SDS-stable apoE/A� levels, theseresults resolve previous contradictory in vitro findings (21, 28,30, 43). HEK-apoE has been utilized in numerous studies forapoE/A� formation (21, 28). Previous data have demonstratedthat HEK-apoE3 but not HEK-apoE4 forms an SDS-stableapoE/A� as measured using Western analysis of SDS-PAGE(21, 28, 30, 43). However, a corresponding value for total com-plex levels was not possible byWestern analysis. With this newELISA, the apoE isoforms exhibit a comparable affinity for A�in the absence of SDS, defined here as total apoE/A�, consistentwith previous reports using nonstringent conditions to mea-sure the complex (31). Themechanism by which the apoE4/A�complex is less stable is unclear. ApoE4/A� disruption canoccur by global effects on protein structure, disrupting thebinding sites on the individual proteins, as well as local effectsat the complex interface. ApoE4 has a lower stability andincreased propensity to populate an intermediate molten glob-ule conformation compared with the other isoforms (55, 56).The apoE4/A� complex exhibits the lowest stability under all

denaturing conditions, potentially due to the greater suscepti-bility of the apoE4 tertiary structure to disruption. Therefore,specific effects on the complex interface cannot be separatedfrom effects on the stability/structure of the individual compo-nents apoE and A�. An additional consideration for the stabil-ity of the apoE/A� is the effect of apoE isoform on lipoproteinlipidation. Increased lipoprotein lipidation increases the levelsof SDS-stable apoE/A�, as determined by Western analysis ofSDS-PAGE (21, 22, 30, 43, 57, 58). If glial cell-derived apoE4 isless lipidated than apoE3, the apoE4/A� complex would be lessstable (59, 60). Thus, the biochemical development and charac-terization of the ELISA have resolved some of the inconsisten-cies in the apoE/A� literature. Having validated the ELISA invitro, we used it in vivo to address the hypothesis that the levelsof soluble apoE/A� isoform specially modulate oA� levels.

Soluble oA� levels are thought to include the proximal neu-rotoxic A� assemblies in AD (61). Soluble oA� assemblies areneurotoxic in vitro and in vivo (61), and both soluble A� andoA� correlate with disease progression inADpatients (62–65).We previously demonstrated that soluble levels of total A�42and oA� were increased in E4FAD transgenic mice comparedwith E2FAD and E3FAD, although the levels of apoEwere com-parable, suggesting a functional difference between the iso-forms. In this study, soluble apoE4/A� complex levels werelower than apoE2/A� and apoE3/A� complex levels in EFADmice. These data indicate an inverse association betweenapoE/A� and oA� levels and are consistent with previous pub-lications that suggest that apoE/A� levels isoform-specificallymodulate soluble A� (11, 15). Synapse degeneration is consid-ered a proximal cause of cognitive deficits in AD. ApoE/A�complex levels may affect synaptic A� levels and function. Atthe synapse, as with the whole brain, apoE/A� appears to bepresent as an insoluble and soluble form. IHC co-localiza-tion of apoE and A� at the synapse is a measure of primarilyinsoluble apoE/A� (35, 36), and data demonstrate insolubleapoE/A� appears to form preferentially with apoE4 com-pared with apoE3 (36). Previous results have shown that thedetergent and guanidine extraction pattern of mouse apoEparallels that of A�42 in 5�FAD mice (5), and the propor-tion of apoE/A� in insoluble fractions was increased in ADsynaptosomes compared with controls.6 Importantly, insolu-ble apoE4/A� complex may accumulate in autophagic struc-tures within synaptic terminals (66, 67). However, the effect ofAPOE genotype on soluble synaptic apoE/A� levels is less clear.Soluble A�, soluble oA�, p-Tau, and SDS-stable p-Tau oligo-mers (37–39, 68) are detected in AD synaptosomes, and datapresented herein also demonstrate the presence of solubleapoE/A� in AD synaptosomes, with levels reduced comparedwith controls. Soluble apoE/A� levels were also lower in synap-tosomes fromADpatients withAPOE4 comparedwithAPOE3.These data indicate a difference in apoE/A� solubility duringdisease progression, which may lead to alterations in synapticA� trafficking or clearance.

Although the cellular process by which the apoE isoformmodulates soluble A� pathology is unclear, a number of apoE/

6 K. H. Gylys, unpublished observations.

FIGURE 6. AD prediction by A�42, oA�, apoE/A�, T-Tau, and p-Tau-181 inhuman CSF using ROC curves. ROC curves for A�42, oA�, apoE/A�, T-Tau,and p-Tau-181 in control and AD patients with the APOE3/3 genotype inhuman CSF are shown. ROC curves represent the predicted probabilities ofbeing an AD case using marginal logistic regression models. Specificity (truenegative rate, the proportion of AD controls correctly predicted) is plotted onthe x axis and sensitivity (the proportion of AD cases correctly predicted) isplotted on the y axis, as calculated based on each subject’s predicted caseprobability being above or below the varying threshold, respectively.

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5922 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

A�-based mechanisms have been proposed. Examples includeeffects on the following: 1) A� oligomerization (69, 70); 2) A�clearance via glia (71–74), neurons (75, 76), and/or the blood-brain barrier (77, 78); 3) enzymatic degradation (57); and 4)drainage via the interstitial fluid (ISF) (16) or perivasculature(79). Furthermore, the dynamic compartmentalization of A� intheCNShas been identified as an important factor in regulatingthe level of soluble A� (80), which may be affected by and/oraffect apoE/A�. Importantly, the levels of A� in each compart-ment affect the equilibrium between compartments, again withfurther modulation by the apoE isoforms. To understand thisprocess, new techniques have been developed to determineapoE and A� turnover via stable isotope-labeling kinetics (81)and microdialysis of ISF (80). In addition, Hong et al. (80) haverecently identified A� in biochemically distinct compartmentsin the brain, including an ISF pool, a TBS-extractable pool, anSDS-extractable pool, and an insoluble or plaque pool. InFAD-Tg mice with a high plaque burden, ISF A� appears to berapidly sequestered in a TBS-soluble pool (80). Overall, themajority of A� in the ISF originates from a less soluble paren-chymal A� pool rather than from production (80). ApoE iso-form-specific apoE/A� levels could affect the dynamic com-partmentalization of A� through the mechanisms discussedabove. For example, with APOE3, high levels of solubleapoE/A� may reduce soluble and oA� levels via clearance.With APOE4, low levels of soluble apoE/A� may result inincreased soluble A� levels, particularly oA�. Alternatively, ifapoE is acting as a pathological chaperone for soluble A�,reducing the level or stability of apoE/A� may decrease oA�levels (24, 82). As described herein, the ability to detect apoEisoform-specific differences in the levels of soluble oA� andapoE/A� levels in vivo is a critical step in identifying the mech-anism by which the apoE isoforms modulate soluble A�pathology.As with human synaptosomes, in human CSF levels of solu-

ble oA� were greater and apoE/A� lower with APOE4 com-pared with APOE3 in the AD cohort. The ability of both oA�and apoE/A� to distinguish between APOE3/3 and APOE4/4ADpatients is consistent with the increased risk and earlier ageof disease onset with APOE4, highlighting the potential forthese markers to track disease progression. In addition,apoE/A� and oA� may represent novel CSF biomarkers, animportant focus for AD research (54). In control and ADpatients with the APOE3/3 genotype, both oA� and apoE/A�diagnosedADwith the same accuracy as p-Tau-181, a currentlyacceptedADbiomarker. Furthermore, as oA� and apoE/A� arebased on potential mechanisms of AD progression, both repre-sent biomarkers to assess therapeutic efficacy in vivo and inclinical trials. Currently, drug trials targeting oA� and apoE/A�are either in the preclinical phase or underway. For A�, thera-pies include both passive and active A� immunotherapy,�-secretase inhibitors, �-secretase inhibitors, and �-secretasemodulators. Aside from measures of cognition, neuroimagingfor amyloid with Pittsburgh compound B and CSF biomarkerssuch asA�42 andp-Tau levels are the only biomarkers availableto determine drug efficacy (83). Given that amyloid plaquesappear not to correlate with dementia and may not representthe ideal target, and it is unclear whether low CSF A�42 levels

will be reversible with long term treatment, the relevance ofthese biomarkers for therapeutic trials is unclear. oA� levelsrepresent a novel biomarker to monitor drug efficacy. ForapoE/A�, therapies are in development that disrupt (24, 82) orincrease (84) apoE/A� levels. Examples include retinoid Xreceptor, liver X receptor, and peroxisome proliferator-acti-vated receptor-� agonists, which increase the levels and lipid-ation state of apoE (57, 84–86), apoE structural correctors (87),andA�12–28P that blocks apoE/A� interactions (24, 82).How-ever, there are no data on whether these drugs will affectapoE/A� levels in vivo, and more importantly, the effects ofthese therapeutic interventions on the human apoE isoformsare unknown. The data presented here indicate that increasingsoluble levels of apoE/A� is a therapeutic target, as it will reduceoA� levels. Importantly the efficacy of therapeutic treatmentstargeting soluble levels of oA� and apoE/A� can now be deter-mined using the ELISAs described herein.

Acknowledgments—Human post-mortem samples were obtainedfrom the Alzheimer’s Disease Research Center Neuropathology Coresof the University of Southern California (supported by National Insti-tutes of Health Grant 050 AG05142 from NIA), UCLA (supported byNational Institutes of Health Grant P50 AG 16970 from NIA), andUniversity of California at Irvine (supported by National Institutes ofHealth Grant P50 AG016573 from NIA). Human CSF samples wereobtained from the University of Kentucky Alzheimer’s DiseaseResearch Center (supported by National Institutes of Health GrantP30 AG028383 from NIA). We also gratefully acknowledge MariaCorrada and Claudia Kawas and the 90� Study for providing tissue(University of California at Irvine).

REFERENCES1. Verghese, P. B., Castellano, J. M., and Holtzman, D. M. (2011) Apolipo-

protein E in Alzheimer’s disease and other neurological disorders. LancetNeurol. 10, 241–252

2. Bu, G. (2009) Apolipoprotein E and its receptors in Alzheimer’s disease.Pathways, pathogenesis, and therapy. Nat. Rev. Neurosci. 10, 333–344

3. Hardy, J. A., and Higgins, G. A. (1992) Alzheimer’s disease. The amyloidcascade hypothesis. Science 256, 184–185

4. Wisniewski, T., and Frangione, B. (1992) Apolipoprotein E. A pathologicalchaperone protein in patients with cerebral and systemic amyloid.Neuro-sci. Lett. 135, 235–238

5. Youmans, K. L., Leung, S., Zhang, J.,Maus, E., Baysac, K., Bu,G., Vassar, R.,Yu, C., and LaDu, M. J. (2011) Amyloid-�42 alters apolipoprotein E solu-bility in brains of mice with five familial ADmutations. J. Neurosci. Meth-ods 196, 51–59

6. Strittmatter,W. J., Saunders, A.M., Goedert,M.,Weisgraber, K. H., Dong,L. M., Jakes, R., Huang, D. Y., Pericak-Vance, M., Schmechel, D., andRoses, A. D. (1994) Isoform-specific interactions of apolipoprotein E withmicrotubule-associated protein Tau. Implications for Alzheimer disease.Proc. Natl. Acad. Sci. U.S.A. 91, 11183–11186

7. Strittmatter, W. J., Saunders, A. M., Schmechel, D., Pericak-Vance, M.,Enghild, J., Salvesen, G. S., and Roses, A. D. (1993) Apolipoprotein E. Highavidity binding to �-amyloid and increased frequency of type 4 allele inlate-onset familial Alzheimer disease. Proc. Natl. Acad. Sci. U.S.A. 90,1977–1981

8. Drzezga, A., Grimmer, T., Henriksen, G.,Muhlau,M., Perneczky, R., Mie-derer, I., Praus, C., Sorg, C., Wohlschlager, A., Riemenschneider, M.,Wester, H. J., Foerstl, H., Schwaiger, M., and Kurz, A. (2009) Effect ofAPOE genotype on amyloid plaque load and gray matter volume in Al-zheimer disease. Neurology 72, 1487–1494

9. Grimmer, T., Tholen, S., Yousefi, B. H., Alexopoulos, P., Forschler, A.,

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5923

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Forstl, H., Henriksen, G., Klunk, W. E., Mathis, C. A., Perneczky, R., Sorg,C., Kurz, A., and Drzezga, A. (2010) Progression of cerebral amyloid loadis associatedwith the apolipoprotein E 4 genotype inAlzheimer’s disease.Biol. Psychiatry 68, 879–884

10. Jones, P. B., Adams, K.W., Rozkalne, A., Spires-Jones, T. L., Hshieh, T. T.,Hashimoto, T., von Armin, C. A., Mielke, M., Bacskai, B. J., and Hyman,B. T. (2011) Apolipoprotein E. Isoform-specific differences in tertiarystructure and interaction with amyloid-� in humanAlzheimer brain. PloSONE 6, e14586

11. Bales, K. R., Liu, F., Wu, S., Lin, S., Koger, D., DeLong, C., Hansen, J. C.,Sullivan, P. M., and Paul, S. M. (2009) Human APOE isoform-dependenteffects on brain �-amyloid levels in PDAPP transgenic mice. J. Neurosci.29, 6771–6779

12. Haass, C., and Selkoe, D. J. (2007) Soluble protein oligomers in neurode-generation. Lessons from the Alzheimer’s amyloid �-peptide. Nat. Rev.Mol. Cell Biol. 8, 101–112

13. Hardy, J. (2006) Alzheimer’s disease. The amyloid cascade hypothesis. Anupdate and reappraisal. J. Alzheimers Dis. 9, 151–153

14. Larson,M. E., and Lesne, S. E. (2012) Soluble A� oligomer production andtoxicity. J. Neurochem. 120, Suppl. 1, 125–139

15. Youmans, K. L., Tai, L. M., Nwabuisi-Heath, E., Jungbauer, L., Kanekiyo,T., Gan, M., Kim, J., Eimer, W. A., Estus, S., Rebeck, G. W., Weeber, E. J.,Bu, G., Yu, C., and Ladu, M. J. (2012) APOE4-specific changes in A�

accumulation in a new transgenic mouse model of Alzheimer disease.J. Biol. Chem. 287, 41774–41786

16. Castellano, J. M., Kim, J., Stewart, F. R., Jiang, H., DeMattos, R. B., Patter-son, B. W., Fagan, A. M., Morris, J. C., Mawuenyega, K. G., Cruchaga, C.,Goate, A. M., Bales, K. R., Paul, S. M., Bateman, R. J., and Holtzman, D.M.(2011) Human apoE isoforms differentially regulate brain amyloid-� pep-tide clearance. Sci. Transl. Med. 3, 89ra57

17. Aleshkov, S., Abraham, C. R., and Zannis, V. I. (1997) Interaction of nas-cent ApoE2, ApoE3, and ApoE4 isoforms expressed in mammalian cellswith amyloid peptide �(1–40). Relevance to Alzheimer’s disease. Bio-chemistry 36, 10571–10580

18. Bentley, N.M., Ladu,M. J., Rajan, C., Getz, G. S., and Reardon, C. A. (2002)Apolipoprotein E structural requirements for the formation of SDS-stablecomplexes with �-amyloid-(1–40). The role of salt bridges. Biochem. J.366, 273–279

19. Golabek, A., Marques, M. A., Lalowski, M., and Wisniewski, T. (1995)Amyloid �-binding proteins in vitro and in normal human cerebrospinalfluid. Neurosci. Lett. 191, 79–82

20. Golabek, A. A., Soto, C., Vogel, T., and Wisniewski, T. (1996) The inter-action between apolipoprotein E and Alzheimer’s amyloid �-peptide isdependent on �-peptide conformation. J. Biol. Chem. 271, 10602–10606

21. LaDu,M. J., Falduto,M. T.,Manelli, A.M., Reardon, C. A., Getz, G. S., andFrail, D. E. (1994) Isoform-specific binding of apolipoprotein E to �-amy-loid. J. Biol. Chem. 269, 23403–23406

22. LaDu, M. J., Lukens, J. R., Reardon, C. A., and Getz, G. S. (1997) Associa-tion of human, rat, and rabbit apolipoprotein E with �-amyloid. J. Neuro-sci. Res. 49, 9–18

23. Pillot, T., Goethals, M., Vanloo, B., Lins, L., Brasseur, R., Vandekerckhove,J., and Rosseneu, M. (1997) Specific modulation of the fusogenic proper-ties of the Alzheimer �-amyloid peptide by apolipoprotein E isoforms.Eur. J. Biochem. 243, 650–659

24. Sadowski, M., Pankiewicz, J., Scholtzova, H., Ripellino, J. A., Li, Y.,Schmidt, S. D., Mathews, P. M., Fryer, J. D., Holtzman, D. M., Sigurdsson,E. M., and Wisniewski, T. (2004) A synthetic peptide blocking the apo-lipoprotein E/�-amyloid binding mitigates �-amyloid toxicity and fibrilformation in vitro and reduces �-amyloid plaques in transgenic mice.Am. J. Pathol. 165, 937–948

25. Yamauchi, K., Tozuka, M., Hidaka, H., Nakabayashi, T., Sugano, M.,Kondo, Y., Nakagawara, A., and Katsuyama, T. (2000) Effect of apolipo-protein AII on the interaction of apolipoprotein E with �-amyloid. Someapo(E-AII) complexes inhibit the internalization of �-amyloid in culturesof neuroblastoma cells. J. Neurosci. Res. 62, 608–614

26. Yang, D. S., Smith, J. D., Zhou, Z., Gandy, S. E., and Martins, R. N. (1997)Characterization of the binding of amyloid-� peptide to cell culture-de-rived native apolipoprotein E2, E3, and E4 isoforms and to isoforms from

human plasma. J. Neurochem. 68, 721–72527. Zhou, Z., Smith, J. D., Greengard, P., and Gandy, S. (1996) Alzheimer

amyloid-� peptide forms denaturant-resistant complex with type 3 butnot type 4 isoform of native apolipoprotein E.Mol. Med. 2, 175–180

28. LaDu, M. J., Pederson, T. M., Frail, D. E., Reardon, C. A., Getz, G. S., andFalduto,M. T. (1995) Purification of apolipoprotein E attenuates isoform-specific binding to �-amyloid. J. Biol. Chem. 270, 9039–9042

29. Strittmatter,W. J.,Weisgraber, K. H., Huang, D. Y., Dong, L.M., Salvesen,G. S., Pericak-Vance, M., Schmechel, D., Saunders, A. M., Goldgaber, D.,and Roses, A. D. (1993) Binding of human apolipoprotein E to syntheticamyloid �-peptide. Isoform-specific effects and implications for late-on-set Alzheimer disease. Proc. Natl. Acad. Sci. U.S.A. 90, 8098–8102

30. LaDu,M. J.,Munson, G.W., Jungbauer, L., Getz, G. S., Reardon, C. A., Tai,L. M., and Yu, C. (2012) Preferential interactions between ApoE-contain-ing lipoproteins and A� revealed by a detection method that combinessize exclusion chromatography with nonreducing gel-shift. Biochim. Bio-phys. Acta 1821, 295–302

31. Morikawa,M., Fryer, J. D., Sullivan, P.M., Christopher, E. A.,Wahrle, S. E.,DeMattos, R. B., O’Dell,M. A., Fagan, A.M., Lashuel, H. A.,Walz, T., Asai,K., and Holtzman, D. M. (2005) Production and characterization of astro-cyte-derived human apolipoprotein E isoforms from immortalized astro-cytes and their interactions with amyloid-�. Neurobiol. Dis. 19, 66–76

32. Manelli, A. M., Stine, W. B., Van Eldik, L. J., and LaDu, M. J. (2004) ApoEand A�1–42 interactions. Effects of isoform and conformation on struc-ture and function. J. Mol. Neurosci. 23, 235–246

33. Russo, C., Angelini, G., Dapino, D., Piccini, A., Piombo, G., Schettini, G.,Chen, S., Teller, J. K., Zaccheo, D., Gambetti, P., and Tabaton, M. (1998)Opposite roles of apolipoprotein E in normal brains and in Alzheimer’sdisease. Proc. Natl. Acad. Sci. U.S.A. 95, 15598–15602

34. Zhou, Z., Relkin, N., Ghiso, J., Smith, J. D., and Gandy, S. (2002) Humancerebrospinal fluid apolipoprotein E isoforms are apparently inefficient atcomplexing with synthetic Alzheimer’s amyloid-� peptide (A�1–40) invitro.Mol. Med. 8, 376–381

35. Arold, S., Sullivan, P., Bilousova, T., Teng, E., Miller, C. A., Poon, W. W.,Vinters, H. V., Cornwell, L. B., Saing, T., Cole, G. M., and Gylys, K. H.(2012) Apolipoprotein E level and cholesterol are associated with reducedsynaptic amyloid � in Alzheimer’s disease and apoE TR mouse cortex.Acta Neuropathol. 123, 39–52

36. Koffie, R. M., Hashimoto, T., Tai, H. C., Kay, K. R., Serrano-Pozo, A.,Joyner, D., Hou, S., Kopeikina, K. J., Frosch, M. P., Lee, V. M., Holtzman,D. M., Hyman, B. T., and Spires-Jones, T. L. (2012) Apolipoprotein E4effects in Alzheimer’s disease are mediated by synaptotoxic oligomericamyloid-�. Brain 135, 2155–2168

37. Henkins, K. M., Sokolow, S., Miller, C. A., Vinters, H. V., Poon, W. W.,Cornwell, L. B., Saing, T., and Gylys, K. H. (2012) Extensive p-Tau pathol-ogy and SDS-stable p-Tau oligomers in Alzheimer’s cortical synapses.Brain Pathol. 22, 826–833

38. Gylys, K. H., Fein, J. A., Yang, F., Wiley, D. J., Miller, C. A., and Cole, G.M.(2004) Synaptic changes in Alzheimer’s disease. Increased amyloid-� andgliosis in surviving terminals is accompanied by decreased PSD-95 fluo-rescence. Am. J. Pathol. 165, 1809–1817

39. Fein, J. A., Sokolow, S., Miller, C. A., Vinters, H. V., Yang, F., Cole, G. M.,andGylys, K. H. (2008) Co-localization of amyloid� and Tau pathology inAlzheimer’s disease synaptosomes. Am. J. Pathol. 172, 1683–1692

40. Gylys, K. H., Fein, J. A., Yang, F., Miller, C. A., and Cole, G. M. (2007)Increased cholesterol in A�-positive nerve terminals from Alzheimer’sdisease cortex. Neurobiol. Aging 28, 8–17

41. Youmans, K. L., Tai, L. M., Kanekiyo, T., Stine, W. B., Jr., Michon, S. C.,Nwabuisi-Heath, E., Manelli, A. M., Fu, Y., Riordan, S., Eimer, W. A.,Binder, L., Bu, G., Yu, C., Hartley, D. M., and LaDu, M. J. (2012) Intraneu-ronal A� detection in 5�FAD mice by a new A�-specific antibody. Mol.Neurodegeneration 7, 8

42. Munson, G. W., Roher, A. E., Kuo, Y. M., Gilligan, S. M., Reardon, C. A.,Getz, G. S., and LaDu,M. J. (2000) SDS-stable complex formation betweennative apolipoprotein E3 and �-amyloid peptides. Biochemistry 39,16119–16124

43. LaDu,M. J., Stine,W. B., Jr., Narita,M., Getz, G. S., Reardon, C. A., and Bu,G. (2006) Self-assembly of HEK cell-secreted ApoE particles resembles

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5924 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

ApoE enrichment of lipoproteins as a ligand for the LDL receptor-relatedprotein. Biochemistry 45, 381–390

44. Dahlgren, K. N., Manelli, A. M., Stine, W. B., Jr., Baker, L. K., Krafft, G. A.,and LaDu, M. J. (2002) Oligomeric and fibrillar species of amyloid-� pep-tides differentially affect neuronal viability. J. Biol. Chem. 277,32046–32053

45. Stine, W. B., Jr., Dahlgren, K. N., Krafft, G. A., and LaDu, M. J. (2003) Invitro characterization of conditions for amyloid-� peptide oligomeriza-tion and fibrillogenesis. J. Biol. Chem. 278, 11612–11622

46. Stine, W. B., Jungbauer, L., Yu, C., and LaDu, M. J. (2011) Preparing syn-thetic A� in different aggregation states.Methods Mol. Biol. 670, 13–32

47. TheNational Institute on Aging, and Reagan InstituteWorking Group onDiagnostic Criteria for the Neuropathological Assessment of Alzheimer’sDisease (1997) Consensus recommendations for the postmortem diagno-sis of Alzheimer’s disease. Neurobiol. Aging 18, S1–S2

48. Nelson, P. T., Braak, H., and Markesbery, W. R. (2009) Neuropathologyand cognitive impairment in Alzheimer disease. A complex but coherentrelationship. J. Neuropathol. Exp. Neurol. 68, 1–14

49. Robin, X., Turck, N., Hainard, A., Tiberti, N., Lisacek, F., Sanchez, J. C.,and Muller, M. (2011) pROC. An open-source package for R and S� toanalyze and compare ROC curves. BMC Bioinformatics 12, 77

50. R Development Core Team (2012) R: A language and environment forstatistical computing. R Foundation for Statistical Computing, Vienna,Austria. ISBN 3–900051-07-0

51. DeLong, E. R., DeLong, D. M., and Clarke-Pearson, D. L. (1988) Compar-ing the areas under two or more correlated receiver operating character-istic curves. A nonparametric approach. Biometrics 44, 837–845

52. Findlay, J. W., and Dillard, R. F. (2007) Appropriate calibration curve fit-ting in ligand binding assays. AAPS J. 9, E260–E267

53. Moore, B. D., Rangachari, V., Tay, W. M., Milkovic, N. M., and Rosen-berry, T. L. (2009) Biophysical analyses of synthetic amyloid-�(1–42) ag-gregates before and after covalent cross-linking. Implications for deducingthe structure of endogenous amyloid-� oligomers. Biochemistry 48,11796–11806

54. Shoji, M. (2011) Biomarkers of the dementia. Int. J. Alzheimers Dis.2011:564321

55. Morrow, J. A., Hatters, D.M., Lu, B., Hochtl, P., Oberg, K. A., Rupp, B., andWeisgraber, K. H. (2002) Apolipoprotein E4 forms a molten globule. Apotential basis for its association with disease. J. Biol. Chem. 277,50380–50385

56. Morrow, J. A., Segall, M. L., Lund-Katz, S., Phillips, M. C., Knapp, M.,Rupp, B., andWeisgraber, K. H. (2000) Differences in stability among thehuman apolipoprotein E isoforms determined by the amino-terminal do-main. Biochemistry 39, 11657–11666

57. Jiang, Q., Lee, C. Y., Mandrekar, S., Wilkinson, B., Cramer, P., Zelcer, N.,Mann, K., Lamb, B., Willson, T. M., Collins, J. L., Richardson, J. C., Smith,J. D., Comery, T. A., Riddell, D., Holtzman, D. M., Tontonoz, P., andLandreth, G. E. (2008) ApoE promotes the proteolytic degradation of A�.Neuron 58, 681–693

58. Hirsch-Reinshagen, V., Maia, L. F., Burgess, B. L., Blain, J. F., Naus, K. E.,McIsaac, S. A., Parkinson, P. F., Chan, J. Y., Tansley, G. H., Hayden, M. R.,Poirier, J., Van Nostrand,W., andWellington, C. L. (2005) The absence ofABCA1 decreases soluble ApoE levels but does not diminish amyloid de-position in two murine models of Alzheimer disease. J. Biol. Chem. 280,43243–43256

59. Gong, J. S., Kobayashi,M.,Hayashi, H., Zou, K., Sawamura,N., Fujita, S. C.,Yanagisawa, K., and Michikawa, M. (2002) Apolipoprotein E (ApoE) iso-form-dependent lipid release from astrocytes prepared from humanApoE3 and ApoE4 knock-in mice. J. Biol. Chem. 277, 29919–29926

60. Riddell, D. R., Zhou, H., Atchison, K., Warwick, H. K., Atkinson, P. J.,Jefferson, J., Xu, L., Aschmies, S., Kirksey, Y., Hu, Y., Wagner, E., Parratt,A., Xu, J., Li, Z., Zaleska, M. M., Jacobsen, J. S., Pangalos, M. N., andReinhart, P. H., (2008) Impact of apolipoprotein E (ApoE) polymorphismon brain ApoE levels. J. Neurosci. 28, 11445–11453

61. Walsh, D. M., and Teplow, D. B. (2012) Alzheimer’s disease and the am-yloid �-protein. Prog. Mol. Biol. Transl. Sci. 107, 101–124

62. Kuo, Y. M., Emmerling, M. R., Vigo-Pelfrey, C., Kasunic, T. C., Kirkpat-rick, J. B., Murdoch, G. H., Ball, M. J., and Roher, A. E. (1996) Water-

soluble A� (N-40, N-42) oligomers in normal and Alzheimer diseasebrains. J. Biol. Chem. 271, 4077–4081

63. Selkoe, D. J. (2011) Resolving controversies on the path to Alzheimer’stherapeutics. Nat. Med. 17, 1060–1065

64. Tomic, J. L., Pensalfini, A., Head, E., and Glabe, C. G. (2009) Soluble fibril-lar oligomer levels are elevated in Alzheimer’s disease brain and correlatewith cognitive dysfunction. Neurobiol. Dis. 35, 352–358

65. Jin, M., Shepardson, N., Yang, T., Chen, G., Walsh, D., and Selkoe, D. J.(2011) Soluble amyloid �-protein dimers isolated from Alzheimer cortexdirectly induce Tau hyperphosphorylation and neuritic degeneration.Proc. Natl. Acad. Sci. U.S.A. 108, 5819–5824

66. Nixon, R. A. (2007) Autophagy, amyloidogenesis, and Alzheimer disease.J. Cell Sci. 120, 4081–4091

67. Li, J., Kanekiyo, T., Shinohara,M., Zhang, Y., Ladu,M. J., Xu,H., andBu,G.(2012) Differential regulation of amyloid-� endocytic trafficking and lys-osomal degradation by apolipoprotein E isoforms. J. Biol. Chem. 287,44593–44601

68. Sokolow, S., Henkins, K. M., Bilousova, T., Miller, C. A., Vinters, H. V.,Poon,W., Cole, G.M., and Gylys, K. H. (2012) AD synapses contain abun-dant A� monomer and multiple soluble oligomers, including a 56-kDaassembly. Neurobiol. Aging 33, 1545–1555

69. Cerf, E., Gustot, A., Goormaghtigh, E., Ruysschaert, J.M., andRaussens, V.(2011) High ability of apolipoprotein E4 to stabilize amyloid-� peptideoligomers, the pathological entities responsible for Alzheimer’s disease.FASEB J. 25, 1585–1595

70. Petrlova, J., Hong, H. S., Bricarello, D. A., Harishchandra, G., Lorigan,G. A., Jin, L. W., and Voss, J. C. (2011) A differential association ofapolipoprotein E isoforms with the amyloid-� oligomer in solution. Pro-teins 79, 402–416

71. Koistinaho,M., Lin, S.,Wu, X., Esterman,M., Koger, D., Hanson, J., Higgs,R., Liu, F., Malkani, S., Bales, K. R., and Paul, S. M. (2004) ApolipoproteinE promotes astrocyte colocalization and degradation of deposited amy-loid-� peptides. Nat. Med. 10, 719–726

72. Mandrekar, S., Jiang, Q., Lee, C. Y., Koenigsknecht-Talboo, J., Holtzman,D. M., and Landreth, G. E. (2009) Microglia mediate the clearance ofsoluble A� through fluid phase macropinocytosis. J. Neurosci. 29,4252–4262

73. Basak, J.M., Verghese, P. B., Yoon, H., Kim, J., andHoltzman, D.M. (2012)Low density lipoprotein receptor represents an apolipoprotein E-inde-pendent pathway of A� uptake and degradation by astrocytes. J. Biol.Chem. 287, 13959–13971

74. Thal, D. R. (2012) The role of astrocytes in amyloid �-protein toxicity andclearance. Exp. Neurol. 236, 1–5

75. Vekrellis, K., Ye, Z., Qiu, W. Q., Walsh, D., Hartley, D., Chesneau, V.,Rosner,M. R., and Selkoe,D. J. (2000)Neurons regulate extracellular levelsof amyloid �-protein via proteolysis by insulin-degrading enzyme. J. Neu-rosci. 20, 1657–1665

76. Wirths, O., and Bayer, T. A. (2012) Intraneuronal A� accumulation andneurodegeneration. Lessons from transgenic models. Life Sci. 91,1148–1152

77. Deane, R., Sagare, A., Hamm, K., Parisi, M., Lane, S., Finn, M. B., Holtz-man, D. M., and Zlokovic, B. V. (2008) ApoE isoform-specific disruptionof amyloid � peptide clearance from mouse brain. J. Clin. Invest. 118,4002–4013

78. Bachmeier, C., Beaulieu-Abdelahad, D., Crawford, F., Mullan, M., andParis, D. (2013) J. Mol. Neurosci., 48, 270–276

79. Hawkes, C. A., Sullivan, P. M., Hands, S., Weller, R. O., Nicoll, J. A., andCarare, R. O. (2012) Disruption of arterial perivascular drainage of amy-loid-� from the brains of mice expressing the human APOE 4 allele. PloSONE 7, e41636

80. Hong, S., Quintero-Monzon, O., Ostaszewski, B. L., Podlisny, D. R., Ca-vanaugh, W. T., Yang, T., Holtzman, D. M., Cirrito, J. R., and Selkoe, D. J.(2011) Dynamic analysis of amyloid �-protein in behaving mice revealsopposing changes in ISF versus parenchymal A� during age-relatedplaque formation. J. Neurosci. 31, 15861–15869

81. Basak, J. M., Kim, J., Pyatkivskyy, Y., Wildsmith, K. R., Jiang, H., Parsadanian,M., Patterson, B. W., Bateman, R. J., and Holtzman, D. M. (2012) Measure-ment of apolipoprotein E and amyloid � clearance rates in the mouse brain

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

FEBRUARY 22, 2013 • VOLUME 288 • NUMBER 8 JOURNAL OF BIOLOGICAL CHEMISTRY 5925

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

using bolus stable isotope labeling.Mol. Neurodegeneration 7, 1482. Sadowski, M. J., Pankiewicz, J., Scholtzova, H., Mehta, P. D., Prelli, F.,

Quartermain, D., and Wisniewski, T. (2006) Blocking the apolipoproteinE/amyloid-� interaction as a potential therapeutic approach for Alzhei-mer’s disease. Proc. Natl. Acad. Sci. U.S.A. 103, 18787–18792

83. Morris, J. C., and Selkoe, D. J. (2011) Recommendations for the incorpo-ration of biomarkers into Alzheimer clinical trials. An overview. Neuro-biol. Aging 32, S1–S3

84. Cramer, P. E., Cirrito, J. R.,Wesson, D.W., Lee, C. Y., Karlo, J. C., Zinn, A. E.,Casali, B. T., Restivo, J. L., Goebel,W.D., James,M. J., Brunden, K. R.,Wilson,D. A., and Landreth, G. E. (2012) ApoE-directed therapeutics rapidly clear�-amyloid and reverse deficits inADmousemodels. Science 335, 1503–1506

85. Lefterov, I., Bookout, A., Wang, Z., Staufenbiel, M., Mangelsdorf, D., andKoldamova, R. (2007) Expression profiling in APP23 mouse brain. Inhibi-tion of A� amyloidosis and inflammation in response to LXR agonisttreatment.Mol. Neurodegeneration 2, 20

86. Terwel, D., Steffensen, K. R., Verghese, P. B., Kummer, M. P., Gustafsson,J. Å., Holtzman, D. M., and Heneka, M. T. (2011) Critical role of astroglialapolipoprotein E and liver X receptor-� expression for microglial A�phagocytosis. J. Neurosci. 31, 7049–7059

87. Mahley, R.W., and Huang, Y. (2012) Small-molecule structure correctorstarget abnormal protein structure and function. Structure corrector res-cue of apolipoprotein E4-associated neuropathology. J. Med. Chem. 55,8997–9008

APOE Genotype Effects on Soluble ApoE/A� Complex Levels

5926 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 288 • NUMBER 8 • FEBRUARY 22, 2013

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from

Hoppens Gylys and Mary Jo LaDuV. Vinters, Linda J. Van Eldik, David W. Fardo, Steve Estus, Guojun Bu, Karen

HarryYoumans, Chunjiang Yu, Wayne W. Poon, Lindsey B. Cornwell, Carol A. Miller, Leon M. Tai, Tina Bilousova, Lisa Jungbauer, Stephen K. Roeske, Katherine L.

Mouse Model and Human Samples and Alzheimer Disease in a TransgenicAPOE4 Increased with βOligomeric A

) Complex Are Reduced andβ (AβLevels of Soluble Apolipoprotein E/Amyloid-

doi: 10.1074/jbc.M112.442103 originally published online January 4, 20132013, 288:5914-5926.J. Biol. Chem. 

  10.1074/jbc.M112.442103Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

Supplemental material:

  http://www.jbc.org/content/suppl/2013/01/04/M112.442103.DC1

http://www.jbc.org/content/suppl/2013/02/21/M112.442103.DCAuthor_profileRead an Author Profile for this article at 

  http://www.jbc.org/content/288/8/5914.full.html#ref-list-1

This article cites 86 references, 28 of which can be accessed free at

by guest on October 1, 2018

http://ww

w.jbc.org/

Dow

nloaded from


Recommended