Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol...

17
Endoplasmic Reticulum Stress Mediates Amyloid b Neurotoxicity via Mitochondrial Cholesterol Trafcking Q68 Elisabet Barbero-Camps,* y Anna Fernández,* y Anna Baulies,* y Laura Martinez,* y Jose C. Fernández-Checa,* yz and Anna Colell* y Q2 Q3 Q4 Q5 From the Department of Cell Death and Proliferation,* Biomedical Research Institute of Barcelona - Higher Council for Scientic Research (IIBB-CSIC), Barcelona, Spain; the Liver Unit, y Hospital Clinic, Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), the Center for Biomedical Research in Hepatic and Digestive Diseases (CIBEREHD), Barcelona, Spain; and the Southern California Research Center for Alcoholic Liver and Pancreatic Diseases, z Keck School of Medicine, University of Southern California, Los Angeles, California Q6 Accepted for publication March 18, 2014. Address correspondence to: Anna Colell, Ph.D., or Jose C. Fernández-Checa, Ph.D., Department of Cell Death and Proliferation, Institut dInvesti- gacions Biomèdiques de Barcelona-CSIC, Rosselló 161, 08036 Barcelona, Spain Q10 . E-mail: [email protected]. es, josecarlos.fernandezcheca@ iibb.csic.es or checa229@ yahoo.com. Disrupted cholesterol homeostasis has been reported in Alzheimer disease and is thought to contribute to disease progression by promoting amyloid b (Ab) accumulation. In particular, mitochondrial cholesterol enrichment has been shown to sensitize to Ab-induced neurotoxicity. However, the mo- lecular mechanisms responsible for the increased cholesterol levels and its trafcking to mitochondria in Alzheimer disease remain poorly understood. Here, we show that endoplasmic reticulum (ER) stress triggered by Ab promotes cholesterol synthesis and mitochondrial cholesterol inux, resulting in mitochondrial glutathione (mGSH Q11 ) depletion in older age Q12 amyloid precursor protein/presenilin-1 (APP/ PS1) mice. Mitochondrial cholesterol accumulation was associated with increased expression of mitochondrial-associated ER membrane proteins, which favor cholesterol translocation from ER to mitochondria along with specic cholesterol carriers, particularly the steroidogenic acute regulatory protein. In vivo treatment with the ER stress inhibitor 4-phenylbutyric acid prevented mitochondrial cholesterol loading and mGSH depletion, thereby protecting APP/PS1 mice against Ab-induced neurotoxicity. Similar protection was observed with GSH ethyl ester administration, which replenishes mGSH without affecting the unfolded protein response, thus positioning mGSH Q13 depletion downstream of ER stress. Overall, these results indicate that Ab-mediated ER stress and increased mitochondrial cholesterol trafcking contribute to the pathologic progression observed in old APP/PS1 mice, and that ER stress inhibitors may be explored as therapeutic agents for Alzheimer disease. (Am J Pathol 2014, -:1e16; http://dx.doi.org/10.1016/j.ajpath.2014.03.014) Since Q14 the initial observation that feeding rabbits with a cholesterol-enriched diet leads to amyloid b (Ab) accumu- lation, 1 the number of studies linking cholesterol and Ab metabolism have increased exponentially. Although the impact of cholesterol in clinical studies remains unsettled, cumulating evidence indicates increased cholesterol levels in brain from Alzheimer disease (AD) patients. 2e5 More- over, ndings in animal and cultured-cell models demon- strate that cholesterol enrichment in lipid rafts fosters Ab production 6e8 and aggregation, 9 and inhibits the intracellular degradation of the peptide. 10 By using mouse models of cholesterol loading, namely sterol regulatory element binding protein 2 (SREBP-2) transgenic mice and Niemann-Pick type C1 knockout mice, we have shown that the trafcking of cholesterol to mitochondria depletes mitochondrial glutathione (mGSH), which in turn exacerbates Ab-induced oxidative neuronal death. 11 Furthermore, in the amyloid precursor protein/presenilin-1 (APP/PS1) transgenic mouse the overexpression of the endoplasmic reticulum (ER)-resi- dent transcription factor SREBP-2 accelerated and worsened key pathologic manifestations of AD, correlating with mitochondrial cholesterol loading and mGSH depletion. 12 Supported in part by Plan Nacional de Investigación Cientíca, Desar- rollo e Innovación Tecnológica Q7 grants SAF2010-15760 (A.C.) and SAF2009-11417 and SAF2012-34831 Q8 (J.C.F.-C.), the Fundació Marató TV3 (J.C.F.-C.), the Instituto de Salud Carlos III grant PI041804 (J.C.F.-C.), the Research Center for Alcoholic Liver and Pancreatic Dis- eases of the NIH/National Institute on Alcohol Abuse and Alcoholism grant P50AA011999-15 (J.C.F.-C.), the Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (A.C. and J.C.F.-C.), and Formación de Personal Investigador (FPI) fellowships from the Ministerio de Economía y Competitividad (E.B.-C., A.B. and L.M.). Copyright ª 2014 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajpath.2014.03.014 ajp.amjpathol.org The American Journal of Pathology, Vol. -, No. -, - 2014 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 FLA 5.2.0 DTD ĸ AJPA1729_proof ĸ 19 May 2014 ĸ 11:53 am ĸ EO: AJP13_0447

Transcript of Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol...

Page 1: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q68

Q2Q3Q4Q5

Q6

Q10

Q14

The American Journal of Pathology, Vol. -, No. -, - 2014

1234567891011121314151617181920212223242526272829303132333435363738394041424344454647484950515253545556575859606162

ajp.amjpathol.org

63646566676869707172737475767778

Endoplasmic Reticulum Stress Mediates Amyloid b

Neurotoxicity via Mitochondrial Cholesterol TraffickingElisabet Barbero-Camps,*y Anna Fernández,*y Anna Baulies,*y Laura Martinez,*y Jose C. Fernández-Checa,*yz and Anna Colell*y

798081828384

From the Department of Cell Death and Proliferation,* Biomedical Research Institute of Barcelona - Higher Council for Scientific Research (IIBB-CSIC),Barcelona, Spain; the Liver Unit,y Hospital Clinic, Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), the Center for Biomedical Research inHepatic and Digestive Diseases (CIBEREHD), Barcelona, Spain; and the Southern California Research Center for Alcoholic Liver and Pancreatic Diseases,z

Keck School of Medicine, University of Southern California, Los Angeles, California

858687

Accepted for publication

11 Q12

C

P

h

8889909192939495969798

March 18, 2014.

Address correspondence to:Anna Colell, Ph.D., or Jose C.Fernández-Checa, Ph.D.,Department of Cell Death andProliferation, Institut d’Investi-gacions Biomèdiques deBarcelona-CSIC, Rosselló 161,08036 Barcelona, Spain.E-mail: [email protected], [email protected] or [email protected].

13

opyright ª 2014 American Society for Inve

ublished by Elsevier Inc. All rights reserved

ttp://dx.doi.org/10.1016/j.ajpath.2014.03.014

99100101102103104105

Disrupted cholesterol homeostasis has been reported in Alzheimer disease and is thought to contributeto disease progression by promoting amyloid b (Ab) accumulation. In particular, mitochondrialcholesterol enrichment has been shown to sensitize to Ab-induced neurotoxicity. However, the mo-lecular mechanisms responsible for the increased cholesterol levels and its trafficking to mitochondriain Alzheimer disease remain poorly understood. Here, we show that endoplasmic reticulum (ER) stresstriggered by Ab promotes cholesterol synthesis and mitochondrial cholesterol influx, resulting inmitochondrial glutathione (mGSH Q) depletion in older age amyloid precursor protein/presenilin-1 (APP/PS1) mice. Mitochondrial cholesterol accumulation was associated with increased expression ofmitochondrial-associated ER membrane proteins, which favor cholesterol translocation from ER tomitochondria along with specific cholesterol carriers, particularly the steroidogenic acute regulatoryprotein. In vivo treatment with the ER stress inhibitor 4-phenylbutyric acid prevented mitochondrialcholesterol loading and mGSH depletion, thereby protecting APP/PS1 mice against Ab-inducedneurotoxicity. Similar protection was observed with GSH ethyl ester administration, which replenishesmGSH without affecting the unfolded protein response, thus positioning mGSH Qdepletion downstreamof ER stress. Overall, these results indicate that Ab-mediated ER stress and increased mitochondrialcholesterol trafficking contribute to the pathologic progression observed in old APP/PS1 mice, and thatER stress inhibitors may be explored as therapeutic agents for Alzheimer disease. (Am J Pathol 2014,-: 1e16; http://dx.doi.org/10.1016/j.ajpath.2014.03.014)

Supported in part by Plan Nacional de Investigación Científica, Desar-rollo e Innovación Tecnológica Q7grants SAF2010-15760 (A.C.) andSAF2009-11417 and SAF2012-34831 Q8(J.C.F.-C.), the Fundació MaratóTV3 (J.C.F.-C.), the Instituto de Salud Carlos III grant PI041804(J.C.F.-C.), the Research Center for Alcoholic Liver and Pancreatic Dis-eases of the NIH/National Institute on Alcohol Abuse and Alcoholism grantP50AA011999-15 (J.C.F.-C.), the Centro de Investigación Biomédica enRed de Enfermedades Hepáticas y Digestivas (A.C. and J.C.F.-C.), andFormación de Personal Investigador (FPI) fellowships from the Ministeriode Economía y Competitividad (E.B.-C., A.B. and L.M.).

106107108109110111112113114115116117118119120121122123

Since the initial observation that feeding rabbits with acholesterol-enriched diet leads to amyloid b (Ab) accumu-lation,1 the number of studies linking cholesterol and Abmetabolism have increased exponentially. Although theimpact of cholesterol in clinical studies remains unsettled,cumulating evidence indicates increased cholesterol levelsin brain from Alzheimer disease (AD) patients.2e5 More-over, findings in animal and cultured-cell models demon-strate that cholesterol enrichment in lipid rafts fosters Abproduction6e8 and aggregation,9 and inhibits the intracellulardegradation of the peptide.10 By using mouse models ofcholesterol loading, namely sterol regulatory element bindingprotein 2 (SREBP-2) transgenic mice and Niemann-Picktype C1 knockout mice, we have shown that the traffickingof cholesterol to mitochondria depletes mitochondrialglutathione (mGSH), which in turn exacerbates Ab-induced

stigative Pathology.

.

FLA 5.2.0 DTD � AJPA1729_proof

oxidative neuronal death.11 Furthermore, in the amyloidprecursor protein/presenilin-1 (APP/PS1) transgenic mousethe overexpression of the endoplasmic reticulum (ER)-resi-dent transcription factor SREBP-2 accelerated and worsenedkey pathologic manifestations of AD, correlating withmitochondrial cholesterol loading and mGSH depletion.12

124

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 2: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q15

Q16 Q17

Q18

19

20

21

22

23

Barbero-Camps et al

125126127128129130131132133134135136137138139140141142143144145146147148149150151152153154155156157158159160161162163164165166167168169170171172173174175176177178179180181182183184185186

187188189190191192193194195196197198199200201202203204205206207208209210211212213214215216217218219220221222223224225226227228229230231232233234235236237238239240241242243244245246247

Consequently, in vivo mGSH recovery in APP/PS1 micesignificantly reduced Tau pathology and Ab depositions.12

Interestingly, increased mitochondrial cholesterol levels,concomitant with reduced mGSH content, also were ob-served in old APP/PS1 mice after Ab accumulation,11 sug-gesting that Ab per se can regulate cellular cholesterol.However, although the role of APP and the amyloidogenicprocessing on cholesterol homeostasis disruption has beensuggested previously,2,13e15 the molecular mechanismsremain largely unknown.

Previous studies have reported that moderate ER stresscan deregulate lipid metabolism.16 The cellular response toalleviate the ER stress is elicited by the stress transducersinositol-requiring protein 1 (IRE1), activating transcriptionfactor 6 (ATF6), and protein kinase R (PRKR)-like endo-plasmic reticulum kinase (PERK) that trigger a signalingpathway called the unfolded protein response (UPR). Inparticular, the engagement of the PERK pathway has beeninvolved in the activation of SREBPs.17e19 Conversely,overexpression of the chaperone glucose regulated protein78 (GRP78) has been described to inhibit the expression ofSREBP-1c and SREBP-2 proteins.20 ER stress is an earlyevent in AD and has been implicated indirectly as a medi-ator of Ab neurotoxicity,21e25 whereas caspase-12 andcaspase-4 have been reported as intermediates of the ERstress-specific apoptosis triggered by Ab.26,27 Hence, thesestudies lead us to hypothesize that Ab-induced ER stressmight be an additional mechanism contributing to theincreased brain cholesterol content observed in AD, as aresult of enhanced SREBP-2 processing. By using 2- to 15-month-old APP/PS1 mice and the SH-SY5Y cell line weexamined the regulatory role of ER stress on cholesterol andmGSH levels and further analyzed the impact of these al-terations on neuronal death. In vivo treatment of APP/PS1mice with ER stress inhibitors restored the mitochondrialcholesterol homeostasis, replenished mGSH, and blockedAb-mediated cell death and neurotoxicity. A similar degreeof protection was observed in APP/PS1 mice treated withGSH ethyl ester, which prevented mGSH depletion withoutaffecting ER stress. These data provide evidence that ERstress contributes to Ab-induced neurotoxicity, at least inpart by increasing mitochondrial cholesterol accumulation,which leads to impaired mitochondrial antioxidant defense.

Materials and Methods

APP/PS1 and SREBP-2 Mice

Breeding pairs of B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J(APP/PS1) and B6;SJLTg(rPEPCKSREBF2)788Reh/J(SREBP-2) mice were purchased from The Jackson Labo-ratory (Bar Harbor, ME) and characterized as previouslydescribed.11 Briefly, at the time of weaning (21 days), micewere identified genetically by PCR using DNA from ear-tipsand following the genotyping protocols provided by thesupplier. We observed an age-dependent increase of brain

2FLA 5.2.0 DTD � AJPA1729_proo

cholesterol levels in female wild-type (WT) mice comparedwith male mice (not shown). Therefore, although femaleAPP/PS1 mice have a more pronounced AD phenotype, inthe present study we focused on male transgenic and WTmice to minimize variability caused by sex differences. Allprocedures involving animals and their care were approvedby the ethics committee of the University of Barcelona andwere conducted in accordance with institutional guidelinesin compliance with national and international laws andpolicies.

Subcellular Fractionation to Isolate Mitochondria, ER,and MAM Purification

Brains were removed of olfactory bulbs, midbrain, andcerebellum, and were homogenated in 210 mmol/Lmannitol, 60 mmol/L sucrose, 10 mmol/L KCl, 10 mmol/Lsodium succinate, 1 mmol/L ADP, 0.25 mmol/L dithio-threitol, 0.1 mmol/L EGTA, and 10 mmol/L HEPES, pH7.4. Homogenates were centrifuged at 700 � g for 10 mi-nutes, and the supernatants were centrifuged at 10,000 � gfor 15 minutes. The resulting pellet (crude mitochondria)was suspended in 2 mL, loaded onto 8 mL of 30% (v/v)Percoll (GE Healthcare Bio-Sciences AB, Uppsala, Sweden) Q

gradient, and centrifuged at 95,000 � g for 30 minutes. Themitochondrial pellet then was rinsed twice by centrifugingfor 15 minutes at 10,000 � g. To isolate mitochondria fromcell culture, 5 � 107 SH-SY5Y cells were trypsinized andrinsed first in PBS and then in hypotonic buffer (20 mmol/LHEPES, 5 mmol/L KCl, and 1.5 mmol/L MgCl2, pH 8.0).After centrifugation at 800 � g for 2 minutes the resultingpellet was resuspended in three Qvolumes of hypotonic bufferand incubated for 10 minutes on ice. Then, cells were dis-rupted by four aspirations through a 30-gauge needle.Mannitol-sucrose-HEPES buffer (2.5�) Q(50 mmol/LHEPES, 525 mmol/L mannitol, 175 mmol/L sucrose, and 5mmol/L EDTA, pH 8.0) was added to the lysates beforecentrifuging at 1500 � g for 5 minutes. The postnuclearsupernatant was separated further through a 14% Percolldensity gradient and centrifuged at 12,000 � g for 20 mi-nutes. Loose mitochondrial pellets were rinsed in 210 mmol/L mannitol, 60 mmol/L sucrose, 10 mmol/L KCl, 10 mmol/L sodium succinate, 0.1 mmol/L EGTA, and 10 mmol/LHEPES, pH 7.4, by centrifuging for 10 minutes at 8000 � g.In both cases (brain and SH-SY5Y cells), mitochondrialpurity was examined for cross-contamination with ER andplasma membrane by Western blot analysis.ER and mitochondria-associated ER membrane (MAM)

were isolated according to the Wieckowski et al28 protocol.Postnuclear supernatant (S1) was centrifuged at 9000 � gfor 10 minutes to further process supernatants Q(S2) andpellets (P2). The ER fraction was obtained by washing theS2 supernatant at 20,000 � g for 15 minutes, followed bycentrifugation at 100,000 � g for 1 hour to collect the pellet.In Qparallel, P2 was centrifuged twice at 10,000 � g beforecentrifugation on a 30% Percoll gradient to obtain the lower

ajp.amjpathol.org - The American Journal of Pathology

248

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 3: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q24

Q25

Q26

Q27

Q28

Q29

Q30

½T1�½T1�

31

32

33

34

35

36

37

38 Q39

40

41

Ab, ER Stress, and Cholesterol Q1

249250251252253254255256257258259260261262263264265266267268269270271272273274275276277278279280281282283284285286287288289290291292293294295296297298299300301302303304305306307308309310

311312313314315316317318319320321322323324325326327328329330331332333334335336337338339340341342343344345346347348349350351352353354355356357358359360361362363364365366367368369370371

band, which contains mitochondria, and the upper band,which contains MAM, which were collected and washedfurther to remove any Percoll contaminants.

Cell Culture and Treatments

The SH-SY5Y human neuroblastoma cell line was obtainedfrom the European Collection of Cell Cultures (Sigma-Aldrich, St. Louis, MO) and grown in Dulbecco’s modifiedEagle’s medium/Ham’s nutrient mixture F12 (Invitrogen,Life Technologies, Carlsbad, CA), containing 10% fetalbovine serum and 2 mmol/L glutamine. Cells were plated ata density of 0.5 � 106 cells/well. Twenty-four hours afterplating, cells were treated with 10 mmol/L oligomeric Ab(Bachem AG, Bubendorf, Switzerland), 2 nmol/L thapsi-gargin (Sigma-Aldrich), 5 mmol/L 4-phenylbutyric acid(PBA) (Enzo Life Sciences, Farmingdale, NY), 50 mmol/Lsalubrinal (Calbiochem, Merck Millipore, Billerica, MA),100 mmol/L tauroursodeoxycholic acid (Calbiochem, MerckMillipore), or 1 nmol/L gonadotropin-releasing hormone(Sigma-Aldrich). In some cases, cells were fractionated intocytosol or mitochondria on digitonin permeabilization of theplasma membrane as described previously.29

Preparation of Ab Peptides

Human Ab1-42 was dissolved to 1 mmol/L in hexafluoro-isopropanol and aliquoted into microcentrifuge tubes, thenthe hexafluoroisopropanol was evaporated and the peptideswere stored at �20�C until use. For oligomeric assembly,concentrated peptides were resuspended in 5 mmol/Ldimethyl sulfoxide and then diluted to 100 mmol/L in phenolredefree media and incubated at 4�C for 24 hours. Oligo-meric forms of Ab were confirmed by Western blot aspreviously described.11

GSH and Cholesterol Measurements

GSH levels in homogenates and mitochondria were analyzedby the recyclingmethod.30 For cholesterol determination frombrain, samples were extracted with alcoholic KOH, distilledwater, and hexane (1:1:2, v/v/v). Appropriate aliquots ofthe hexane layer were evaporated and used for cholesterolmeasurement.31 High-performance liquid chromatographyanalysis was performed using a mBondapak C18 10 mmreverse-phase column (30 cm� 4 mm inner diameter; WatersCorp., Milford, MA), with a mobile phase of 2-propanol/acetonitrile/distilled water (6:3:1, v/v/v) at a flow rate of 1mL/min. The amount of cholesterol was calculated from standardcurves and the identity of the peaks was confirmed by spikingthe sample with known standards. Cholesterol levels inmembrane fractions fromERandMAMwerequantifiedby theAmpex Red cholesterol assay (Invitrogen). Samples wereresuspended in the homogenization buffer containing 5%Triton X-100 (Sigma-Aldrich) and briefly sonicated. Mito-chondrial cholesterol levels in cells were determined after Ab

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

exposure in the presence of the 0.75 mmol/L P450 side-chaincleavage enzyme (CYP11A1) inhibitor aminoglutethimide.

Western Blot Analysis

Samples (30 to 80 mg of protein per lane) were resolved bySDS-PAGE and transferred to nitrocellulose membranes.Blots were probed with the antibodies listed in Table 1.After overnight incubation at 4�C, bound antibodies werevisualized using horseradish-peroxidaseecoupled secondaryantibodies and an ECL developing kit ( QGE Healthcare Bio-Science AB, Uppsala, Sweden). Densitometry of the bandswas measured with QuantityOne Qsoftware (Bio-Rad Labora-tories, Hercules, CA) and values were normalized to b-actinor glyceraldehyde-3-phosphate dehydrogenase.

Immunohistochemistry

Paraffin-embedded blocks were prepared by sequentialdehydration in graded ethanol and infiltration in paraffinbefore embedding. Blocks were sectioned serially at athickness of 5 mm from �1.2 mm through �2.4 mm fromBregma Q. Sections were processed according to the avidinebiotineperoxidase staining method (Vectastain ABC kit;Vector QLaboratories, Burlingame, CA). After antigenretrieval, the endogenous peroxidase was blocked by expo-sure to 3% H2O2 in methanol for 30 minutes. Samples thenwere treated with the avidin Q/biotin blocking kit (VectorLaboratories) according to the manufacturer’s instructionsand incubated overnight at 4�C with mouse monoclonal anti-ATF6 (1:500; Acris, San Diego, CA) and mouse monoclonalantieAb1-42 (1:300; Sigma-Aldrich Q). After washing withPBS, sections were incubated for 1 hour with a biotinylatedsecondary antibody (1:400), followed by ABC staining for1 hour. The immunoreaction was visualized with dia-minobenzidine using a diaminobenzidine-enhanced liquidsubstrate system (Sigma-Aldrich). Sections were counter-stained with hematoxylin (Dako, Glostrup, Denmark).TUNEL labeling was performed using the in situ Cell DeathDetection Kit (Roche Applied Science, Penzberg, Germany).

Immunofluorescence and Laser Confocal Imaging

Dewaxed hippocampal sections were first boiled in citratebuffer (10 mmol/L sodium citrate, pH 6.0) and incubatedwith 0.1 mol/L glycine/PBS for 20 minutes to reduce auto-fluorescence. The sections were incubated overnight at 4�Cwith rabbit polyclonal anti-steroidogenic acute regulatoryprotein (StAR; 1:500; Abcam Q, Cambridge, UK), mousemonoclonal anti-microtubule-associated protein 2 (MAP2;1:500; Sigma-Aldrich Q), and rat monoclonal antieglial fibril-lary acidic protein (GFAP, 1:500; Calbiochem Merck Milli-pore). After washing in PBS, antibody stainingwas visualizedusing Alexa Fluor Q488 and 594 secondary antibodies (1:400;Invitrogen, Life Technologies). Confocal images werecollected using a Leica TCS QSPE laser scanning confocal

3

372

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 4: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q42

Q43

½T2�½T2�

44

45

Q73

Table 1 Details of the Primary Antibodies Used in This Study

Antibody Source and type Company Dilution

ABCA1 Rabbit polyclonal Novus Biologicals (Littleton, CO) 1:1000Calregulin (H-170) Rabbit polyclonal Santa Cruz Biotech 1:500Caspase-3 Rabbit polyclonal Cell Q71Signaling (Danvers, MA) 1:1000Caspase-12 Rat monoclonal Sigma-Aldrich 1:500CHOP Mouse monoclonal Cell Signaling 1:1000COX IV Rabbit polyclonal Cell Signaling 1:1000Cytochrome c (clone 7H8.2C12) Mouse monoclonal Santa Cruz Biotech 1:500Phospho-eIF2a Rabbit polyclonal Invitrogen 1:1000eIF2a Mouse monoclonal Abcam 1:1000GAPDH (6C5) Mouse monoclonal Santa Cruz Biotech 1:5000GRP78 Rabbit polyclonal Stressgen (Enzo Life Science) 1:1000HXK I (G-1) Mouse monoclonal Santa Cruz Biotech 1:200HMG-CoA reductase Goat polyclonal Santa Cruz Biotech 1:500INSIG-1 Rabbit polyclonal Abcam 1:1000MAP2 Mouse monoclonal Sigma-Aldrich Q721:500MFN2 Mouse monoclonal Abcam 1:1000StARD3 Rabbit polyclonal Abcam 1:1000Naþ,KþATPase a1 Mouse monoclonal Santa Cruz Biotech 1:500PACS-2 Goat polyclonal Santa Cruz Biotech 1:200Phospho-PERK (Thr980) Rabbit polyclonal Cell Signaling 1:1000PERK Rabbit polyclonal Sigma-Aldrich 1:1000Rab5A Rabbit polyclonal Santa Cruz Biotech 1:250Rab7 Goat polyclonal Santa Cruz Biotech 1:250Sigma receptor (B-5) Mouse monoclonal Santa Cruz Biotech 1:200SREBP-2 Rabbit polyclonal Abcam 1:1000StAR Mouse monoclonal Abcam 1:1000VDAC Rabbit polyclonal Calbiochem (EMD Millipore) 1:500b-actin Mouse monoclonal Sigma-Aldrich 1:10,000

GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MFN2, mitofusin-2; PACS-2, phosphofurin acidic cluster sorting protein 2; VDAC, voltage-dependentanion-selective channel protein.

Barbero-Camps et al

373374375376377378379380381382383384385386387388389390391392393394395396397398399400401402403404405406407408409410411412413414415416417418419420421422423424425426427428429430431432433434

435436437438439440441442443444445446447448449450451452453454455456457458459460461462463464465466467468469470471472473474475476477478479480481482

microscope equippedwithUV excitation, an argon laser, a 63/1.32 OIL PH3 CS objective, and a confocal pinhole set at 1Airy unit (Leica Microsystems GmbH, Wetzlar, Germany).All of the confocal images shown were single optical sec-tions. Immunofluorescence intensities and colocalizationcoefficients were determined using MBF ImageJ softwareversion 1.43m (NIH, Bethesda, MD).

Real-Time RT-PCR

Real-time RT-PCR amplification was performed using theiScript one-step RT-PCR kit (Bio-Rad Laboratories) with

Table 2 Details of the Primers Used in This Study

Name Host Forward sequence

GRP78 H 50-AATGACCAGAATCGCCTGAC-CHOP H 50-GCGCATGAAGGAGAAAGAAC-SREBP2 H 50-TGCCATTGGCCGTTTGTGTC-STAR H 50-ATCTTTTCCGATCTTCTGC-3Stard4 M 50-GAGAGATGGCTGACCCTGAGAStard5 M 50-GACGCGTCGGGCTGG-30

Xbp1 M 50-GGATCTCTAAAACTAGAGGCT

H, human; M, Mus musculus.

4FLA 5.2.0 DTD � AJPA1729_proo

SYBR Green (Molecular Probes, Life Technologies). Theprimer sequences used are listed in Table 2. PCRs were run induplicate for each sample. The relative gene expression wasquantified by the DDCt method.

Analysis of Xbp1 QmRNA Splicing

RNA was reverse-transcribed using Xbp1-specific primers,described in Table 2, that amplified a 600-bp cDNA productencompassing the IRE1 cleavage sites.32 This fragmentwas digested further by PstI Q(New England BioLabs,Ipswich, MA) to show a restriction site that is lost after

Reverse sequence

30 50-CGCTCCTTGAGCTTTTTGTC-30

30 50-ACCATTCGGTCAATCAGAGC-30

3 50-CCCTTCAGTGCAACGGTCATTCAC-300 50-CTTGGGCATCCTTAGCAAC-30

-30 50-TCAGACAGTCCTTCCAGTTTGATC-30

50-AACTCCTCAGATGGCCTCCA-30

TGGTG-3 50-AAACAGAGTAGCAGCGCAGACTGC-30

ajp.amjpathol.org - The American Journal of Pathology

483484485486487488489490491492493494495496

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 5: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q46

½F1�½F1�

Ab, ER Stress, and Cholesterol

497498499500501502503504505506507508509510511512513514515516517518519520521522523524525526527528529530531532533534535536537538539540541542543544545546547548549550551552553554555556557558

559560561562563564565566567568569570571572573574575576577578579580581582583584585586587588

IRE1-mediated cleavage and mRNA splicing. The PCRproducts were resolved on a 2% agarose gel.

Pregnenolone Determination

To assess pregnenolone production, cells were plated inDulbecco’s modified Eagle’s medium phenol-free mediacontaining 5% fetal bovine serum and 2 mmol/L glutamine.Cells were exposed to 10 mmol/L Ab with or without pre-treatment (1 hour) with ER stress inhibitors. After 48 hours ofincubation, new media containing 10 mmol/L of trilostane(Santa Cruz Biotechnology, Inc., Santa Cruz, CA) was addedto prevent further processing of pregnenolone, and cells wereleft to accumulate it for 48 hours. Media then was recoveredand levels of the steroid hormone precursor were determinedusing a human pregnenolone enzyme-linked immunosorbentassay kit (Diagnostics Biochem Canada, Inc., Dorchester,ON, Canada), according to the manufacturer’s instructions.Data were normalized to total protein values.

Statistics

Results are expressed as means � SD of the number ofexperiments. Statistical significance was examined using theunpaired, two-tailed Student’s t-test or one-way analysis ofvariance with the Dunnett or Bonferroni post hoc multiplecomparisons test when required. P < 0.05 value wasconsidered statistically significant.

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

Results

Enhanced SREBP-2 Processing and DecreasedATP-Binding Cassette, Sub-Family A, Member 1Transporter Expression Correlate with Increased BrainMitochondrial Cholesterol Levels in APP/PS1 Mice

We first monitored the age-dependent changes in braincholesterol levels of APP/PS1 mice. Cholesterol contentin homogenate or isolated mitochondria from brains ofAPP/PS1 mice significantly increased at 10 months of age(Supplemental Figure S1A), consistent with previous ob-servations.11 The increment in the sterol levels wasobserved in hippocampus samples from 7-month-old APP/PS1 mice (Figure 1A), whereas cholesterol levels in cere-bellum, a brain area relatively unaffected in AD, remainedunchanged at 10 months of age (Figure 1A). These findingswere accompanied by a significant depletion of GSH inbrain mitochondria of 10-month-old APP/PS1 mice(Supplemental Figure S1B). Western blot analysis in thefinal mitochondrial fraction from 10-month-old WT andAPP/PS1 mice showed an absence of plasma membrane andendosome markers, as well as a negligible presence ofPERK and calreticulin compared with ER fraction.Thus, the contribution from cross-contamination of otherorganellar fractions to the higher level of mitochondrialcholesterol seen in aged APP/PS1 mice was minimized(Supplemental Figure S2).

Figure 1 Expression levels of cholesterolbiosynthesis-related proteins. A: Cholesterollevels. Q57B: Western blot analysis of SREBP-2 proteinlevels in brain extracts from WT mice and APP/PS1(A/P) mice at the indicated ages. C: Western blotanalysis of SREBP-2 and HMG-CoA reductase(HMGCR) proteins in brain extracts from 10-month-old WT mice and APP/PS1 mice atdifferent ages. D: Representative immunoblots ofINSIG-1 and ABCA1 presence in brain extractsfrom WT mice and APP/PS1 mice at the indicatedages. Relative intensity values are densitometricvalues of the bands representing the specificprotein immunoreactivity normalized with thevalues of the corresponding b-actin bands.*P < 0.05, **P < 0.01 versus WT mice values byunpaired, two-tailed Student’s t-test (A, B, and D)and one-way analysis of variance with the Dunnettpost hoc test (C) (n Z 4 to 6). mth, month.

5

589590591592593594595596597598599600601602603604605606607608609610611612613614615616617618619620

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 6: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q47

Q48

½F2�½F2�

49

Barbero-Camps et al

621622623624625626627628629630631632633634635636637638639640641642643644645646647648649650651652653654655656657658659660661662663664665666667668669670671672673674675676677678679680681682

683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738739740741742743

To relate these observations with pathways leading tocholesterol synthesis, we analyzed the processing of matureSREBP-2. The increase in the levels of the transcriptionallyactive fragment of SREBP-2 (68 kDa) was first detectable at4 months of age (Figure 1, B and C) and correlated with anenhanced expression of its target gene encoding the enzyme3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA)reductase, the rate-limiting enzyme of the cholesterol syn-thesis pathway (Figure 1C). The expression of SREBP-2 andHMG-CoA reductase further increased in older APP/PS1mice (Figure 1C), whereas the levels of both peptidesremained unchanged in WT mice at all ages analyzed(Supplemental Figure S3A). Because SREBP activation istightly regulated by levels of insulin-induced gene-1 (INSIG-1)17,33 we next analyzed its expression in APP/PS1 mice. Asshown, transgenic mice compared with WT mice displayeddecreased levels of INSIG-1 at all ages analyzed (Figure 1D).Moreover, because of the time lag between the early SREBP-2 processing (age, 4 mo) and the late increase in cholesterollevels (age, 10 mo) observed in APP/PS1 mice, we consid-ered the contribution of alternative mechanisms in regulatingcholesterol homeostasis. For instance, decreased levels of theATP-binding cassette transporter, sub-family A, member 1(ABCA1) have been described in brains of 11-month-oldAPP/PS1 mice, with accumulation of cholesterol in culturedastrocytes resulting from Ab-induced down-regulation ofABCA1 expression.34 In turn, data from mouse models andin vitro experiments have suggested that ABCA1 may affectAb deposition and clearance by regulating apolipoprotein Elipidation and brain cholesterol homeostasis.35 Furthermore,despite the fact that the expression levels of this protein in ADstill are under debate, with studies showing high levels ofABCA1 in hippocampal tissues from AD patients,36,37

several reports have linked ABCA1 polymorphisms to anincreased risk for AD.35 Interestingly, when the levels of theprotein were analyzed in APP/PS1 mice (Figure 1D), weobserved an increase in the expression at 4 and 7 months ofage that decreased to WT levels in 10-month-old mice. Theexistence of an impaired cholesterol efflux resulting from lowABCA1 levels associated with an enhanced synthesis mayexplain in part the cholesterol increase in old APP/PS1 mice.

Increased Expression of Mitochondrial CholesterolCarriers in APP/PS1 Mice

Mitochondria are cholesterol-poor organelles. However,cholesterol transport to the inner mitochondrial membrane(IMM) plays a physiological role and is a highly regulatedprocess.38 Cholesterol trafficking to IMMs is controlled chieflyby StAR, which moves cholesterol from the outer mitochon-drial membrane to the IMM formetabolism. StAR belongs to afamily of proteins that contain StAR-related lipid transfer do-mains (StAR-related lipid transfer proteins), of which StARD3(MLN64) and StARD4 also have been described to allow de-livery of cholesterol to mitochondria. InWTmice the levels ofStAR and StARD3 were maintained steadily during aging

6FLA 5.2.0 DTD � AJPA1729_proo

(Supplemental Figure S3B). By contrast, APP/PS1 mice dis-played high levels of StARD3 at all ages analyzed comparedwith WT mice (Figure 2A), whereas StAR increased in 10-month-old transgenic mice (Figure 2A), concomitant to theincrease of mitochondrial cholesterol observed at this age(Supplemental Figure S1A). The mRNA expression ofStARD4 also was increased in 10-month-old APP/PS1 mice,whereas StARD5, which is involved in the transport ofcholesterol between Golgi, ER, and plasma membrane,remained unchanged (Figure 2B). Immunohistochemicalanalysis by confocal microscopy confirmed the increase ofStAR in the hippocampus of 10-month-old APP/PS1 mice(Figure 2C). The double-labeling showed colocalization be-tween StAR and the neuronal marker anti-MAP2, but not withthe astrocyte marker glial fibrillary acidic protein (Figure 2, Cand D). Similarly, significant up-regulation of StAR andStARD3 protein levels was observed in SREBP-2 transgenicmice (Figure 2E), establishing a link between inducedcholesterol synthesis andmitochondrial cholesterol trafficking.Furthermore, differences in the time expression of bothcholesterol carriers betweenAPP/PS1 and TgeSREBP-2micehighly suggest that although StARD3 may be regulated by thetranscription factor SREBP-2, the induction of StAR expres-sion requires high levels of cholesterol, but is not directlydependent on SREBP-2 activation.In addition to StAR-related lipid transfer proteins as medi-

ators of mitochondrial cholesterol transport, recent studieshave suggested the involvement of MAMs. MAMs are mo-lecular hubs that regulate several aspects of mitochondrialbiology, ranging from calcium transmission to lipid meta-bolism,39 including cholesterol homeostasis.40,41 Moreover,data from experimental AD models have shown increasedMAM function as determined by cholesteryl ester and phos-pholipid synthesis,42 accompanied by high levels of MAM-associated proteins, including the Qsigma-1 receptor,43 andsuggest that these interaction sites may contribute to AD pa-thology. To evaluate the role of MAMs on the enhancedmitochondrial cholesterol transport observed in old APP/PS1mice, we first testedwhether cholesterol enrichment in isolatedMAMs was similar to that of mitochondria. As previouslyreported, cholesterol levels were higher in MAM fractionscompared with the total content in the ER (Figure 2F). How-ever, we did not find significant differences between WT andAPP/PS1 mice in either ER or in the MAM subdomains(Figure 2F). As indicators of increased area of apposition be-tween ER and mitochondria we next evaluated the levels ofphosphofurin acidic cluster sorting protein 2 and mitofusin-2,both are proteins that are required to regulate the contact sitesbetween the two organelles.44,45 As shown, Western blotanalysis showed similar levels of both peptides in MAMsisolated from brain extract of WT and APP/PS1 mice(Figure 2G). Conversely, the levels of the sigma-1 receptor andvoltage-dependent anion-selective channel protein wereincreased significantly in MAMs from APP/PS1 mice(Figure 2G), suggesting that these MAM-associated proteins,previously proposed to facilitate cholesterol translocation from

ajp.amjpathol.org - The American Journal of Pathology

744

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 7: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

print&web4C=FPO

Figure 2 Expression levels of cholesterol-transporting polypeptides. A: Representative immunoblots of StARD3 and StAR showing an increased presence ofthese cholesterol carriers in brain samples from APP/PS1 mice compared with 10-month-old WT mice. B: mRNA levels of StARD4 Q58and StARD5 in brain samplesfrom WT and APP/PS1 mice at 10 months of age analyzed by quantitative RT-PCR. Absolute mRNA values were determined, normalized to 18S rRNA, andreported as relative levels compared to the expression Q59in WT mice. C: Representative confocal images of StAR (red) and MAP2 (green) immunofluorescencelabeling of hippocampal sections from 10-month-old WT mice and APP/PS1 mice. D: Representative confocal images of hippocampal sections from 10-month-old APP/PS1 mice immunostained with anti-StAR (red) and anti-MAP2 or antieglial fibrillary acidic protein (GFAP; green). The extent of colocalization wasquantified using the linear Pearson correlation coefficient (rp); the resulting scatterplot images are shown on the right. A value of 1.0 indicates completecolocalization of two fluorescent signals. Note that StAR preferentially colocalizes with neurons labeled with anti-MAP2 compared with astrocytes labeled withanti-GFAP. E: Western blot analysis of StARD3 and StAR expression in brain from 10-month-old WT mice and TgeSREBP-2 mice at the indicated ages. F: Totalcholesterol in ER and MAMs from brain extracts of 10-month-old WT and APP/PS1 mice. G: Representative immunoblots showing the expression of calreticulin(calregulin), the sigma-1 receptor (s1R), phosphofurin acidic cluster sorting protein 2 (PACS-2), and the mitochondrial proteins cyclooxygenase 4 (COX Q60IV),voltage-dependent anion-selective channel protein (VDAC), and mitofusin 2 (MFN2) in ER, MAMs, and mitochondria from 10-month-old WT and APP/PS1 (A/P)mice. In all Western blot analyses Q61the densitometric values of the bands representing the specific protein immunoreactivity were normalized with the values ofthe corresponding b-actin bands or Q62Ponceau Staining. *P < 0.05, **P < 0.01 versus WT mice values by one-way analysis of variance with the Dunnett post hoctest (A and E) and the unpaired, two-tailed Student’s t-test (B, C, and G) (n Z 3 to 6). Scale bars: 15 mm (C and D). mito., mitochondria; mth, month.

Ab, ER Stress, and Cholesterol

745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806

807808809810811812813814815816817818819820821822823824825826827828829830831832833834835836837838839840841842843844845846847848849850851852853854855856857858859860861862863864865866867

ER to mitochondria,41,46 may contribute to mitochondrialcholesterol content in APP/PS1 mice.

Early ER Stress Precedes Mitochondrial CholesterolAccumulation in APP/PS1 Mice

Given the cause-and-effect relationship between increasedcholesterol synthesis and enhancedmitochondrial cholesterol

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

trafficking and mGSH regulation, we aimed to analyzefurther the mechanism responsible for these changes.Because previous studies had shown that moderate ER stressmodulates the cellular lipid content,16 we explored whetherthe Ab-induced ER stress can affect cholesterol homeostasis.Although no signs of ER stress were detectable inWTmice atall of the ages analyzed (Supplemental Figure S4A), the UPRwas activated in APP/PS1 mice, with markers showing age-

7

868

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 8: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

½F3�½F3�

Q50

½F4�½F4�

WT 4-mth 7-mth 10-mth GRP78

CHOP

p-PERK

APP/PS1

β-actin 0

2

4

6

GRP78CHOP

4-mth 7-mth 10-mthAPP/PS1

WTytisnetni

evit al eR

ATF6APP/PS1WT

**

*

p-eIF2α

p-PERKp-eIF2α

020406080

WT APP/PS1 TUN

1pbXdecil ps

%,

WT APP/PS1TUN

S

U

Unspliced (480 bp)Spliced (454 bp)

PstI

*

7-mth 10-mth 7-mth 10-mth GRP78

Tg-SREBP-2

β-actin

p-eIF2α

0

1

2

3 GRP78ytisnetnievit al e

R

p-eIF2α*

**

WT

7-mth 10-mth 7-mth 10-mth Tg-SREBP-2WT

**

** *

*eIF2α

eIF2α

PERK

(78 kDa)

(27 kDa)

(78 kDa)

(170 kDa)

(125 kDa)

(40 kDa)

(40 kDa)

(40 kDa)

(40 kDa)

Figure 3 APP/PS1 mice display early ER stress.A: Representative immunoblots showing theexpression of the chaperonic protein GRP78 andthe ER stress signaling pathway proteins PERK,phospho-PERK, eIF2a, phospho-eIF2a, and CHOPin brain extracts from 10-month-old WT mice andAPP/PS1 mice at the indicated ages. B: Immuno-histochemical staining of ATF6. Representativephotomicrographs of CA1 hippocampus showingnuclear localization of ATF6 in 7-month-old APP/PS1 mice (n Z 3). C: Analysis of Xbp1 mRNAsplicing. Total RNAs from brain of 7-month-old WTand APP/PS1 mice were subjected to RT-PCR asdescribed in Materials and Methods. After diges-tion with PstI, the PCR products were resolved in a2% agarose gel electrophoresis. The PCR productsof Xbp1 mRNA spliced (S) remained intact (454bp), whereas the unspliced products (U) were cutinto two fragments of 289 and 191 bp, as indi-cated by the arrows. mRNA from liver of WT micetreated i.p. with 1 mg/kg 24 hours tunicamycin(TUN Q63) was used as a positive control (n Z 3). D:Western blot analysis of GRP78, eIF2a, andphospho-eIF2a in brain extracts from WT mice andSREBP-2 transgenic mice. In all Western blot an-alyses Q64, densitometric values of the bands repre-senting the specific protein immunoreactivity werenormalized with the values of the correspondingb-actin bands. *P < 0.05 versus WT mice values byone-way analysis of variance with the Dunnettpost hoc test (A), and the unpaired two-tailedStudent’s t-test (D) (n Z 6). Scale bar Z 50mmol/L (B). mth, month.

print&web4C=FPO

Barbero-Camps et al

869870871872873874875876877878879880881882883884885886887888889890891892893894895896897898899900901902903904905906907908909910911912913914915916917918919920921922923924925926927928929930

931932933934935936937938939940941942943944945946947948949950951952953954955956957958959960961962963964965966967968969970971972973974975976977978979980981982983984985986987988989990991

dependent changes (Figure 3A). GRP78 and phospho-PERKwere the earliest markers up-regulated at 4 months of age(Figure 3A and Supplemental Figure S4B), whereas eukary-otic initiation factor (eIF)-2a phosphorylation and C/EBPhomologous protein (CHOP) increase were detected in olderAPP/PS1 mice (Figure 3A). Moreover, Ab loading paralleledER stress in APP/PS1 mice, with hippocampal amyloidaccumulation in intracellular compartments of 4-month-oldAPP/PS1mice (Supplemental Figure S5). APP/PS1mice alsoshowed activation of the ATF6-dependent branch of UPR.ATF6 is processed by the same metalloproteases that activateSREBPs47 and although previous reports suggested thatATF6 antagonizes the activity of SREBP-2,48 we foundnuclear ATF6 in hippocampal neurons from 7-month-oldAPP/PS1 mice (Figure 3B). In contrast, compared withtunicamycin-treated mice, IRE1 phosphorylation and Xbp1splicing were not observed in APP/PS1 mice (data not shownand Figure 3C). The correlation between ER stress andcholesterol accumulation also was noticed in SREPB-2transgenic mice, which showed increased GRP78 levels andeIF2a phosphorylation compared withWTmice (Figure 3D).

8FLA 5.2.0 DTD � AJPA1729_proo

Thus, APP/PS1 mice showed an early UPR response thatpreceded the increase of cholesterol levels, the increase in theexpression of mitochondrial cholesterol transporting poly-peptides, particularly StAR, and mGSH depletion.

Ab Induces ER Stress in SH-SY5Y Cells, Resulting inIncreased Mitochondrial Cholesterol Influx and mGSHDepletion

Once we established that ER stress precedes the disruption incholesterol homeostasis in APP/PS1 mice, we analyzed thelink between ER stress and mitochondrial cholesterol loadingby Ab in vitro. We used soluble oligomeric forms of Ab1-42,previously confirmed by Western blot.11 As shown, Ab1-42exposure increased the expression ofGRP78 andCHOP at themRNA (Figure 4A) and protein levels (Figure 4B) over timein SH-SY5Y cells. These findings were accompanied byeIF2a phosphorylation (Figure 4B), confirming the involve-ment of the PERK branch observed in APP/PS1 mice.This outcome preceded the increased expression of tran-

scription factor SREBP-2 and StAR by Ab both at the

ajp.amjpathol.org - The American Journal of Pathology

992

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 9: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Figure 4 ER stress induced by oligomeric Ab1-42 promotes mitochondrial cholesterol transport and mGSH depletion in SH-SY5Y cells. GRP78,p-eIF2a, and CHOP expression levels in cells treated with 10 mmol/L Ab at the indicated time points, analyzed by quantitative RT-PCR (A) and Westernblot (B). SREBP-2 and StAR expression levels after exposure to 10 mmol/L Ab at the indicated time points, analyzed by quantitative RT-PCR (C) andWestern blot (D). E: mRNA levels of SREBP-2 Q65and StAR after incubation with 2 mmol/L thapsighargin (TG) at the indicated time points, analyzed byquantitative RT-PCR. mRNA values were normalized to 18S rRNA and reported as relative levels compared to the expression Q66in WT mice. F: Mitochondrialcholesterol levels after incubation with 10 mmol/L Ab for 48 hours, and 0.75 mmol/L inhibitor aminoglutethimide was added for the last 24 hours. G:Quantification of pregnenolone delivered to media as indicator of mitochondrial cholesterol loading. Cells were exposed to 10 mmol/L Ab with orwithout 5 mmol/L PBA, 50 mmol/L salubrinal (SAL), or 100 mmol/L tauroursodeoxycholic acid (TUDCA) pretreatment for 1 hour. After 48 hours ofincubation new media containing 20 mmol/L of trilostane was replaced and cells were left to accumulate the steroid hormone for 48 hours. Preg-nenolone levels in media were determined as described in Materials and Methods. As a positive control, cells were treated with 1 nmol/L gonadotropin-releasing hormone (GnRH) for 48 hours. Western blot analysis of the mitochondrial cholesterol carriers StAR and StARD3 in cellular extracts (H) andmitochondrial GSH levels after 4 days of 10 mmol/L Ab incubation with or without 5 mmol/L PBA, 50 mmol/L SAL, or 100 mmol/L TUDCA pretreatmentfor 1 hour (I). In all of the Western blot analyses Q67densitometric values of the bands representing the specific protein immunoreactivity were normalizedwith the values of the corresponding b-actin bands. *P < 0.05 versus control values; yP < 0.05, yyP < 0.01 versus Ab treatment values by one-wayanalysis of variance with the Dunnett post hoc test (AeE), the unpaired, two-tailed Student’s t-test (F), and one-way analysis of variance with theBonferroni multiple comparisons test (G-I) (n Z 3 to 6).

Ab, ER Stress, and Cholesterol

The American Journal of Pathology - ajp.amjpathol.org 9

9939949959969979989991000100110021003100410051006100710081009101010111012101310141015101610171018101910201021102210231024102510261027102810291030103110321033103410351036103710381039104010411042104310441045104610471048104910501051105210531054

10551056105710581059106010611062106310641065106610671068106910701071107210731074107510761077107810791080108110821083108410851086108710881089109010911092109310941095109610971098109911001101110211031104110511061107110811091110111111121113111411151116

FLA 5.2.0 DTD � AJPA1729_proof � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 10: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q51

½F5�½F5�

Q52

½F6�½F6�

Barbero-Camps et al

11171118111911201121112211231124112511261127112811291130113111321133113411351136113711381139114011411142114311441145114611471148114911501151115211531154115511561157115811591160116111621163116411651166116711681169117011711172117311741175117611771178

1179118011811182118311841185118611871188118911901191119211931194119511961197119811991200120112021203120412051206120712081209121012111212121312141215121612171218121912201221122212231224122512261227122812291230123112321233123412351236123712381239

mRNA and protein levels (Figure 4, C and D). Similarfindings were observed when SH-SY5Y cells were exposedto the ER stressor thapsigargin (Figure 4E). Consistent withthe function of StAR in mitochondrial cholesterol traf-ficking, there was a significant increase in cholesterol levelsin isolated mitochondria from SH-SY5Y cells treated withAb for 48 hours (Figure 4F), pointing to the amyloid peptideas responsible for the mitochondrial cholesterol enrichmentobserved in APP/PS1 mice.

To further assess the involvement of the Ab-induced ERstress we tested the effects of ER stress inhibitors. In ste-roidogenic cells, cholesterol is metabolized in the IMM byP450 side-chain cleavage enzyme (CYP11A) and convertedinto the steroid precursor pregnenolone. Mitochondrialcholesterol availability is the rate-limiting step in the steroidbiosynthesis and, therefore, pregnenolone levels can be usedas a gauge of the rate of mitochondrial cholesterol influx.49

In agreement with the increased mitochondrial cholesterollevels observed in isolated mitochondria, exposure to 10mmol/L Ab1-42 resulted in a significant increase in preg-nenolone levels (Figure 4G). This increase was similar tothat caused by gonadotropin-releasing hormone (Figure 4G),which has been described previously to modulate steroido-genesis in SH-SY5Y cells.50 Interestingly, pre-incubation withdifferent chemical inhibitors of ER stress, including 3 mmol/LPBA, 50 mmol/L salubrinal, or 100 mmol/L tauroursodeox-ycholic acid, significantly decreased the levels of StARD3 andStAR protein induced by Ab (Figure 4H) and prevented theincrease of pregnenolone in Ab-exposed cells (Figure 4G). Asexpected for mitochondrial cholesterol loading, Ab challengedepleted mGSH levels (Figure 4I), and this effect was pre-vented by PBA. Thus, these findings establish a cause-and-effect relationship between ER stress triggered by Ab andmGSH depletion.

ER Stress Contributes to in Vivo Neurotoxicity in APP/PS1 Mice due to mGSH Depletion

Although UPR is considered a protective mechanism, it caninitiate an apoptotic cascade if the response persists overtime. To analyze the contribution of ER stress to neuronaldeath we tested the role of PBA treatment in vivo (500 mg/kgper day), administered i.p. every 12 hours for 2 weeks, in 15-month-old APP/PS1 mice, when neurodegeneration is fullydetectable. Treatment with the chaperone significantlyreduced the levels of the ER stress markers GRP78 andCHOP in APP/PS1 mice (Figure 5A). Moreover, the pro-teolytic processing of caspase-12 and caspase-3 to theiractive fragments was blocked by PBA treatment (Figure 5B),resulting in reduced apoptosis in the hippocampus of APP/PS1 mice (Figure 5C). The use of the chaperone-like mole-cule also prevented the expression of SREBP-2 as well as theincrease of the mitochondrial cholesterol carriers StAR andStARD3 observed in APP/PS1 mice (Figure 5D). Parallelingthese effects, mitochondrial cholesterol loading (Figure 5E)and mGSH depletion (Figure 5F) in APP/PS1 mice were

10FLA 5.2.0 DTD � AJPA1729_proo

abolished by PBA therapy, further validating the link be-tween ER stress and mGSH regulation.We previously showed that the mGSH content is critical in

regulating the cellular response to different apoptotic stimuliincluding tumor necrosis factor-a, hypoxia, or Ab peptides.51

Therefore, it is conceivable to speculate that the protectiveeffect exerted by PBA may be owing, at least in part, to therecovery of the pool of mGSH. To further test this hypothesismice were administered 1.25 mmol/kg/d GSH ethyl ester(GSHee) i.p. every 12 hours for 2 weeks. As shown, GSHeereplenished the pool of mGSH (Figure 6A). The expressionof the cholesterol-related proteins SREBP-2 and StARD3, aswell as the increased levels of the ER stress markers GRP78,CHOP, and cleaved caspase-12, observed in APP/PS1 miceremained unchanged after GSHee treatment (Figure 6, B andC). However, GSHee significantly reduced the cleavage ofcaspase-3 (Figure 6D) and prevented brain apoptosis in APP/PS1 mice to a similar extent as observed with PBA(Figure 5C). Overall, these findings suggest that ER stresscontributes to in vivo Ab neurotoxicity in APP/PS1 mice, atleast in part by depletion of mGSH.

Discussion

A recent lipidomic analysis described a complex landscape oflipid alterations in specificbrain regions inAD.52Among these,disruption of cholesterol homeostasis has been described inAD,2e5 and our recent studies advanced a role for mitochon-drial cholesterol in Ab-induced neuroinflammation andneurotoxicity.11,12 Here, we analyzed the role of ER stress incholesterol homeostasis, mitochondrial cholesterol loading,and mGSH depletion in APP/PS1 mice (Figure 6E). First, weobserved that APP/PS1 mice show early ER stress andincreased levels of active SREBP-2 associated with decreasedexpression of INSIG-1. INSIGs exert their action in the ER bybinding SREBP cleavage-activating protein and preventing itfrom escorting SREBPs to the Golgi apparatus where theSREBPs are processed to their active forms. Interestingly,INSIG-1 is a short-lived protein, and inhibition of proteintranslation processes as a response to ER stress has been shownto reduce its levels severely,17 thus raising the possibility that asimilar mechanism may take place in APP/PS1 mice, likelycontributing to enhanced processing of SREBP-2.Despite the presence of active SREBP-2 since 4 months

of age, cholesterol accumulates only in old transgenic mice,and is first detectable in the hippocampus. A potential factorthat accounts for this age-dependent difference may beABCA1 expression, which increases in young transgenicmice but decreases in old mice. Although in-depth researchwould be needed to discern the exact mechanism, chronicinflammatory conditions41,42 or changes in cerebral oxy-sterols53,54 might account for this age-dependent decrease ofABCA1 expression.A key feature of our findings is the increased mitochon-

drial cholesterol flux that correlates with depleted mGSHcontent. The evidence for this outcome was generated in a

ajp.amjpathol.org - The American Journal of Pathology

1240

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 11: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

β-actin

GRP78

CHOP

Caspase-12

APP/PS1

+ PBA

APP/PS1

WT

Caspase-3

WT

APP/PS1

WT+PBA WT+GSHee

APP/PS1+PBA APP/PS1+GSHee

WT +

PBA

E F

WTPBA - + - +

APP/PS1

0

1

2

3ytisnetni .l eR

CHOPGRP78

β-actin

WT+

PBA

WT

APP/PS1

APP/PS1

+ PBA

55 kDa

42 kDaβ-actin

WT+

PBA

WT APP/P

S1APP/P

S1+ P

BA

35 kDa17 kDa

(78 kDa)

(27 kDa)

**

SREBP-2

StAR

β-actin

StARD3

0

2

4

6ytisnetnievit al e

R

StARStARD3

SREBP-2ABP+TWAPP/PS1

**

*

WT APP/PS1

(68 kDa)

(32 kDa)

(50 kDa)

0

2

4

6

GSH

, nm

ol/m

g pr

ot.

*

WT+

PBA

APP/PS1

WT

APP/PS1

+ PBA

WT+

PBAC

hole

ster

ol, μ

g/m

g pr

ot. *

APP/PS1

WT

APP/PS1

+ PBA

0

5

10

15

20

PBA - - +

0

20

40

60

80

100

NT PBA GSHee

APP/PS1WT

**TU

NEL

pos

itive

cel

ls (%

)

01

234ytisnetni .l e

R

Csp12(42 kDa)

*

01

234ytisnetni .l e

R

Csp3(17 kDa) *

WT APP/PS1PBA - + - +

WT APP/PS1PBA - + - +

† † ††

†† ††

†††

print&web4C=FPO

Figure 5 Treatment with the chemical chaperone PBA restores cholesterol homeostasis and provides protection against ER stresseinduced cell death inAPP/PS1 mice. Fifteen-month-old mice were treated i.p. with 500 mg/kg/d PBA for 2 weeks (n Z 6 mice per group) and levels of ER stress markers, caspaseactivation, and cholesterol-related proteins were analyzed by Western blot. A: Representative immunoblots of GRP78 and CHOP presence in brain extracts. B:Representative immunoblots of caspase-12 and caspase-3 cleavage; arrows show fragmented caspase-12 (Csp12) (w42 kDa) and cleaved caspase-3 activefragment (Csp3) (17 kDa). C: Representative images of apoptotic cells in hippocampus from 15-month-old WT and APP/PS1 mice with or without PBA or GSHee(i.p. 1.25 mmol/kg/d for 2 weeks) assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). D: Representative immu-noblots showing a significant reduction of SREBP-2, StARD3, and StAR in brain samples from APP/PS1 mice after PBA treatment. Mitochondrial cholesterolcontent (E) and mGSH levels (F). In all Western blot analyses Q69densitometric values of the bands representing the specific protein immunoreactivity werenormalized with the values of the corresponding b-actin bands. *P < 0.05, **P < 0.01 versus WT values; yP < 0.05, yyP < 0.01 versus APP/PS1 mice withouttreatment by one-way analysis of variance with the Bonferroni multiple comparisons test (n Z 3 to 6). Scale bar Z 50 mmol/L (C).

Ab, ER Stress, and Cholesterol

12411242124312441245124612471248124912501251125212531254125512561257125812591260126112621263126412651266126712681269127012711272127312741275127612771278127912801281128212831284128512861287128812891290129112921293129412951296129712981299130013011302

1303130413051306130713081309131013111312131313141315131613171318131913201321132213231324132513261327132813291330133113321333133413351336133713381339134013411342134313441345134613471348134913501351135213531354135513561357135813591360136113621363

dual fashion. First, the increase in mitochondrial cholesterolparallels that of pregnenolone in human neuroblastomacells challenged with Ab peptide. Because mitochondrialcholesterol availability is the rate-limiting step in steroido-genesis,49,55 pregnenolone determination is a surrogate ofmitochondrial cholesterol trafficking to IMM. Second, we

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

examined the expression of different protein carriers thatpotentially involved in cholesterol transport to mitochon-dria. Unlike StARD3, changes in the expression of StARwere detectable in 10-month-old APP/PS1 mice, parallelingthe increase of cholesterol in mitochondria, which positionsStAR as most likely responsible for mitochondrial

11

1364

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 12: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Figure 6 In vivo GSH ethyl ester treatment. WT and APP/PS1 mice were treated i.p. with 1.25 mmol/kg/d GSHee for 2 weeks (n Z 6 mice per group). A:mGSH levels. B: Representative immunoblots showing the expression levels of SREBP-2 protein and the mitochondrial cholesterol carrier StARD3 in brainextracts after GSHee treatment. C: Western blots analysis of ER stress markers GRP78 and CHOP levels and the presence of cleaved caspase-12 active fragment(42 kDa) in brain extracts after GSHee treatment. D: Representative immunoblots of caspase-3 cleavage; arrow shows cleaved active fragment (17 kDa).Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) levels were analyzed as a loading control. In all Western blot analyses Q70densitometric, values of the bandsrepresenting the specific protein immunoreactivity were normalized with the values of the corresponding GAPDH bands. E: Scheme illustrating the proposedmodel by which Ab-induced ER stress would contribute to neuronal death through promoting cholesterol synthesis and translocation to mitochondria. Thiscascade of events leading to neuronal death could be counteracted by inhibiting ER stress or by restoring the mGSH levels with GSHee therapy. *P < 0.05,**P < 0.01 versus WT mice values; yP < 0.05 versus APP/PS1 mice values by one-way analysis of variance with the Bonferroni multiple comparisons test;n Z (3 to 6). CHOL, cholesterol; Mito., mitochondrial; MMP, mitochondrial membrane permeabilization.

Barbero-Camps et al

13651366136713681369137013711372137313741375137613771378137913801381138213831384138513861387138813891390139113921393139413951396139713981399140014011402140314041405140614071408140914101411141214131414141514161417141814191420142114221423142414251426

1427142814291430143114321433143414351436143714381439144014411442144314441445144614471448144914501451145214531454145514561457145814591460146114621463146414651466146714681469147014711472147314741475147614771478147914801481148214831484148514861487

cholesterol loading. Interestingly, the immunohistologicanalyses show that the increase in StAR is limited to hip-pocampal neurons; although we cannot exclude that anenhanced cholesterol synthesis also may occur in astrocytes,

12FLA 5.2.0 DTD � AJPA1729_proo

which are known to be the primary site for cholesterolloading in adult brain. The relevance of the mitochondrialcholesterol loading described in APP/PS1 mice is reinforcedfurther by the reported presence of highStAR levels in affected

ajp.amjpathol.org - The American Journal of Pathology

1488

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 13: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q53

54

Ab, ER Stress, and Cholesterol

14891490149114921493149414951496149714981499150015011502150315041505150615071508150915101511151215131514151515161517151815191520152115221523152415251526152715281529153015311532153315341535153615371538153915401541154215431544154515461547154815491550

1551155215531554155515561557155815591560156115621563156415651566156715681569157015711572157315741575157615771578157915801581158215831584158515861587158815891590159115921593159415951596159715981599160016011602160316041605160616071608160916101611

brains ofADpatients.56 In these studies, the enrichment inStARwas observed both in hippocampal pyramidal neurons and as-trocytes. Moreover, although recent studies indicate thatStARD3 regulates mitochondrial cholesterol in the absence ofNiemann-Pick type C1,49 it is likely that the differential local-ization of StARD3 (endolysosomal) versus StAR (mitochon-dria) determines their contribution to mitochondrial cholesterolregulation. In addition, posttranslationalmodifications of StAR,including its phosphorylation at serine residues, have beendescribed to modulate steroidogenesis.57 The impact of thesefactors in promotingmitochondrial cholesterol accumulation inthe APP/PS1 mouse model remains to be established.

Mitochondrial cholesterol trafficking implies two events:cholesterol transfer to the outer mitochondrial membranefrom extramitochondrial sources and intramitochondrialcholesterol movement from the outer mitochondrial mem-brane to IMM for metabolism. StAR plays an essential rolein the latter. Indeed, germline StAR deficiency in mice re-sults in lethal congenital adrenal lipoid hyperplasia,58

meaning that other StAR-related lipid transfer memberscannot replace StAR deficiency. A remaining question,however, is how does cholesterol transfer to the outermitochondrial membrane to be available for StAR? Wefound increased levels of the sigma-1 receptor and voltage-dependent anion-selective channel protein, both MAM-associated proteins, which have been shown to interactand collaborate with StAR in the transfer of cholesterol fromER to mitochondria.41,46 The responsiveness of MAM toER stress has been described in Ca2þ-dependent mito-chondrial apoptosis59; in particular, the sigma-1 receptor hasbeen reported to regulate ER/mitochondrial Ca2þ mobili-zation under ER stress conditions.60 Moreover, in additionto these possibilities, caveolin-1 has recently been shown toregulate mitochondrial cholesterol homeostasis.55 However,its putative contribution to Ab-mediated mitochondrialcholesterol loading remains to be investigated. On the otherhand, overexpression of StARD4 in mouse hepatocytes alsohas been reported to stimulate bile acid synthesis,61 sup-porting its role of cholesterol transport to mitochondria.Furthermore, by using cholesterol-containing liposomes asdonors, a recent study62 showed that recombinant StARD4accelerates cholesterol transfer to isolated liver mitochon-dria. Consistent with these studies, we found a 1.5- to 2-foldincrease of Stard4 mRNA levels in 10-month-old APP/PS1mice whereas Stard5 levels remained unchanged, consistentwith its inability to transfer cholesterol to mitochondria.63,64

Previous data have indicated that StARD5 expression wasup-regulated by ER stress63; however, the transcriptionalinduction of StARD5 is mediated by XBP1,65 a signaltransducer that was unprocessed and therefore inactive inthe ER stress response displayed by APP/PS1 mice.

We observed an early activation of the UPR, coincidingwith the burst of Ab generation in 4-month-old APP/PS1mice, and the induction of cholesterol synthesis, indicatingthat Ab-induced ER stress can play a role in this alteration.In support of this link, we observed that Ab-induced ER

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

stress in SH-SY5Y cells stimulates the expression ofSREBP-2 and StAR. The ER stressor thapsigargin repro-duced these findings, confirming and extending the initialobservations describing a link between ER stress andcholesterol synthesis.16,66 Moreover, Ab exposure resulted inaccumulation ofmitochondrial cholesterol and pregnenolone,indicating that Ab promotes cholesterol flux to mitochondria.The efficacy of different ER stress inhibitors in normalizingthe expression of StARD3 and StAR and reducing the hor-mone levels, further proved that this effect was mediated viaER stress. Consistent with these data, we recently providedcompelling evidence in hepatocytes that StAR is an ER stresstarget gene whose expression, induced by tunicamycin, isprevented by tauroursodeoxycholic acid.67 Finally, weobserved that the Ab-enhanced mitochondrial cholesterolinflux is accompanied by significant mGSH depletion. Thelink between these events, namely increased cholesterol andmGSH depletion, is owing to the sensitivity of the mGSHcarrier to changes in membrane dynamics induced bycholesterol accumulation inmitochondrial membranes.68 Thereduction of mGSH levels was prevented by the chemicalchaperone PBA, suggesting a key role of Ab-induced ERstress in controlling the mitochondrial antioxidant defense.

APP/PS1 mice display specific UPR signaling, charac-terized by selective PERK and ATF6 activation, withoutIRE1a phosphorylation and Xbp1 processing. Although theunderlying mechanisms contributing to this particular UPRbranch activation are unknown, initial observations havedescribed that mutations in PS1 increased the vulnerabilityto ER stress owing to a down-regulation of the UPR, causedby disturbed function of the IRE1a.69 Whether this partic-ular UPR signaling is a specific feature of APP/PS1 micehas yet Qto be confirmed.

ER stressemediated cell death has been linked consistentlyto Ab neurotoxicity.21,23,24 Consistent with these findings,APP/PS1 mice showed increased processing of caspase-12and caspase-3 associated with neuronal death, which wasreduced significantly by inhibiting ER stress. Moreover,in vivo treatment with PBA also normalized the expression ofSREBP-2 and that of StAR and StARD3 in APP/PS1 mice,preventing mitochondrial cholesterol accumulation. Recentstudies in Tg2576 and APP/PS1 mice show that chronicadministration of PBA is accompanied by clearance of Ab,reduced Tau pathology, and reversal of learning deficits.70e72

In addition to its established role as an anti-ER stress agent,PBA has been shown to inhibit histone deacetylases, whichmay contribute to PBA-mediated effects by restoring thetranscription of proteins involved in synaptic plasticity andstructural remodeling. Histone deacetylase inhibitors alsohave been described to modulate cholesterol homeostasis,favoring cholesterol trafficking and degradation.73e75 How-ever, the fact that other ER stress inhibitors such as salubrinaland tauroursodeoxycholic acid can modulate cholesterol ho-meostasis minimizes the potential contribution of histonedeacetylase inhibition by PBA, further supporting an ERstressedependent mechanism.

13

1612

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 14: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q55

Barbero-Camps et al

16131614161516161617161816191620162116221623162416251626162716281629163016311632163316341635163616371638163916401641164216431644164516461647164816491650165116521653165416551656165716581659166016611662166316641665166616671668166916701671167216731674

1675167616771678167916801681168216831684168516861687168816891690169116921693169416951696169716981699170017011702170317041705170617071708170917101711171217131714171517161717171817191720172117221723172417251726172717281729173017311732173317341735

Weobserved a significant recovery of themGSHcontent incells and APP/PS1 mice that was associated with PBA-induced cholesterol normalization. Given the key role ofmGSH in controlling the oxidative stress generated in mito-chondria,51 we inferred that part of the ER stressemediatedcell death in the transgenic mice resulted from a compromisedmitochondrial antioxidant defense that sensitizes cells to Abinsult. To confirm this hypothesis, mice were administeredGSHee, which significantly recovered the mitochondrial poolof GSH in APP/PS1 mice. As expected, the treatment did notmodify the levels of ER stress or the increased brain choles-terol content displayed by the transgenic mice, but signifi-cantly reduced caspase-3 cleavage and neuronal death, to thesame extent as PBA treatment, highlighting the key role ofmGSH in regulating ER stressemediated neuronal death. It isworth noting that TgeSREBP-2 mice also showed ER stressassociated with enhanced mitochondrial cholesterol loadingand depletedmGSH levels,11whereas no signs of apoptosis orAb accumulation were observed in thesemice during aging,12

indicating that altered cholesterol homeostasis per se is notsufficient to initiate the disease.

Taken together, our findings point to ER stress as a linkbetween Ab and cholesterol up-regulation and subsequentmitochondrial trafficking (Figure 6E). Moreover, althoughthe relative contribution of neuronal and glial cells has to beestablished, the histochemical analyses show an increasedpresence of StAR and nuclear ATF6 in the hippocampalpyramidal cell layer of transgenic mice, indicating thatderegulated cholesterol homeostasis and ER stress is presentin neurons. In addition, the data uncover mGSH depletion as akey mechanism of ER stress-mediated neurotoxicity in APP/PS1 mice. This view is of particular relevance given recentfindings suggesting a feed-forward loop between ER stressand Ab processing in AD,76 with the expected consequencesof mitochondrial cholesterol loading and mGSH depletioncontributing to Ab-mediated neurotoxicity. Thus, therapeuticstrategies targeting ER stress and resulting in the restorationof mGSH may be of significance in the treatment of AD.

Acknowledgments

We thank Drs. Carmen Garcia-Ruiz, Montserrat Mari, andAlbert Morales for critical comments and suggestions andSusana Nuñez for her help with mice care.

This work was developed, in part, at the Esther KoplowitzCenter (Barcelona, Spain).

Supplemental Data

Supplemental material for this article can be found athttp://dx.doi.org/10.1016/j.ajpath.2014.03.014.

References

1. Sparks DL, Scheff SW, Hunsaker JC 3rd, Liu H, Landers T, Gross DR:Induction of Alzheimer-like beta-amyloid immunoreactivity in the

14FLA 5.2.0 DTD � AJPA1729_proo

brains of rabbits with dietary cholesterol. Exp Neurol 1994, 126:88e94

2. Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A,Hatanpaa K, Troncoso JC, Mattson MP: Involvement of oxidativestress-induced abnormalities in ceramide and cholesterol metabolismin brain aging and Alzheimer’s disease. Proc Natl Acad Sci U S A2004, 101:2070e2075

3. Bandaru VV, Troncoso J, Wheeler D, Pletnikova O, Wang J,Conant K, Haughey NJ: ApoE4 disrupts sterol and sphingolipidmetabolism in Alzheimer’s but not normal brain. Neurobiol Aging2009, 30:591e599

4. Xiong H, Callaghan D, Jones A, Walker DG, Lue LF, Beach TG,Sue LI, Woulfe J, Xu H, Stanimirovic DB, Zhang W: Cholesterolretention in Alzheimer’s brain is responsible for high beta- andgamma-secretase activities and Abeta production. Neurobiol Dis2008, 29:422e437

5. Panchal M, Loeper J, Cossec JC, Perruchini C, Lazar A, Pompon D,Duyckaerts C: Enrichment of cholesterol in microdissected Alz-heimer’s disease senile plaques as assessed by mass spectrometry.J Lipid Res 2010, 51:598e605

6. Osenkowski P, Ye W, Wang R, Wolfe MS, Selkoe DJ: Direct andpotent regulation of gamma-secretase by its lipid microenvironment.J Biol Chem 2008, 283:22529e22540

7. Kalvodova L, Kahya N, Schwille P, Ehehalt R, Verkade P,Drechsel D, Simons K: Lipids as modulators of proteolytic activity ofBACE: involvement of cholesterol, glycosphingolipids, and anionicphospholipids in vitro. J Biol Chem 2005, 280:36815e36823

8. Ehehalt R, Keller P, Haass C, Thiele C, Simons K: Amyloidogenicprocessing of the Alzheimer beta-amyloid precursor protein dependson lipid rafts. J Cell Biol 2003, 160:113e123

9. Yip CM, Elton EA, Darabie AA, Morrison MR, McLaurin J:Cholesterol, a modulator of membrane-associated Abeta-fibrillo-genesis and neurotoxicity. J Mol Biol 2001, 311:723e734

10. Lee CY, Tse W, Smith JD, Landreth GE: Apolipoprotein E promotesbeta-amyloid trafficking and degradation by modulating microglialcholesterol levels. J Biol Chem 2012, 287:2032e2044

11. Fernandez A, Llacuna L, Fernandez-Checa JC, Colell A: Mitochon-drial cholesterol loading exacerbates amyloid beta peptide-inducedinflammation and neurotoxicity. J Neurosci 2009, 29:6394e6405

12. Barbero-Camps E, Fernandez A, Martinez L, Fernandez-Checa JC,Colell A: APP/PS1 mice overexpressing SREBP-2 exhibit combinedAbeta accumulation and tau pathology underlying Alzheimer’s dis-ease. Hum Mol Genet 2013, 22:3460e3476

13. Grimm MO, Grimm HS, Patzold AJ, Zinser EG, Halonen R,DueringM, Tschape JA, De Strooper B,Muller U, Shen J, Hartmann T:Regulation of cholesterol and sphingomyelin metabolism by amyloid-beta and presenilin. Nat Cell Biol 2005, 7:1118e1123

14. Liu Q, Zerbinatti CV, Zhang J, Hoe HS, Wang B, Cole SL, Herz J,Muglia L, Bu G: Amyloid precursor protein regulates brain apoli-poprotein E and cholesterol metabolism through lipoprotein receptorLRP1. Neuron 2007, 56:66e78

15. Pierrot N, Tyteca D, D’Auria L, Dewachter I, Gailly P, Hendrickx A,Tasiaux B, Haylani LE, Muls N, N’Kuli F, Laquerriere A,Demoulin JB, Campion D, Brion JP, Courtoy PJ, Kienlen-Campard P, Octave JN: Amyloid precursor protein controls choles-terol turnover needed for neuronal activity. EMBO Mol Med 2013, 5:608e625

16. Colgan SM, Hashimi AA, Austin RC: Endoplasmic reticulum stressand lipid dysregulation. Expert Rev Mol Med 2011, 13:e4

17. Lee JN, Ye J: Proteolytic activation of sterol regulatory element-binding protein induced by cellular stress through depletion ofInsig-1. J Biol Chem 2004, 279:45257e45265

18. Bobrovnikova-Marjon E, Hatzivassiliou G, Grigoriadou C,Romero M, Cavener DR, Thompson CB, Diehl JA: PERK-dependentregulation of lipogenesis during mouse mammary gland developmentand adipocyte differentiation. Proc Natl Acad Sci U S A 2008, 105:16314e16319

ajp.amjpathol.org - The American Journal of Pathology

1736

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 15: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Ab, ER Stress, and Cholesterol

17371738173917401741174217431744174517461747174817491750175117521753175417551756175717581759176017611762176317641765176617671768176917701771177217731774177517761777177817791780178117821783178417851786178717881789179017911792179317941795179617971798

1799180018011802180318041805180618071808180918101811181218131814181518161817181818191820182118221823182418251826182718281829183018311832183318341835183618371838183918401841184218431844184518461847184818491850185118521853185418551856185718581859

19. Colgan SM, Tang D, Werstuck GH, Austin RC: Endoplasmic retic-ulum stress causes the activation of sterol regulatory element bindingprotein-2. Int J Biochem Cell Biol 2007, 39:1843e1851

20. Kammoun HL, Chabanon H, Hainault I, Luquet S, Magnan C,Koike T, Ferre P, Foufelle F: GRP78 expression inhibits insulin andER stress-induced SREBP-1c activation and reduces hepatic steatosisin mice. J Clin Invest 2009, 119:1201e1215

21. Lee JH, Won SM, Suh J, Son SJ, Moon GJ, Park UJ, Gwag BJ:Induction of the unfolded protein response and cell death pathway inAlzheimer’s disease, but not in aged Tg2576 mice. Exp Mol Med2010, 42:386e394

22. Hoozemans JJ, van Haastert ES, Nijholt DA, Rozemuller AJ,Scheper W: Activation of the unfolded protein response is an earlyevent in Alzheimer’s and Parkinson’s disease. Neurodegener Dis2012, 10:212e215

23. Ferreiro E, Resende R, Costa R, Oliveira CR, Pereira CM: Anendoplasmic-reticulum-specific apoptotic pathway is involved inprion and amyloid-beta peptides neurotoxicity. Neurobiol Dis 2006,23:669e678

24. Umeda T, Tomiyama T, SakamaN, Tanaka S, LambertMP, KleinWL,Mori H: Intraneuronal amyloid beta oligomers cause cell death viaendoplasmic reticulum stress, endosomal/lysosomal leakage, andmitochondrial dysfunction in vivo. J Neurosci Res 2011, 89:1031e1042

25. Chafekar SM, Hoozemans JJ, Zwart R, Baas F, Scheper W: Abeta 1-42 induces mild endoplasmic reticulum stress in an aggregation state-dependent manner. Antioxid Redox Signal 2007, 9:2245e2254

26. Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, Yuan J:Caspase-12 mediates endoplasmic-reticulum-specific apoptosis andcytotoxicity by amyloid-beta. Nature 2000, 403:98e103

27. Hitomi J, Katayama T, Eguchi Y, Kudo T, Taniguchi M, Koyama Y,Manabe T, Yamagishi S, Bando Y, Imaizumi K, Tsujimoto Y,Tohyama M: Involvement of caspase-4 in endoplasmic reticulumstress-induced apoptosis and Abeta-induced cell death. J Cell Biol2004, 165:347e356

28. Wieckowski MR, Giorgi C, Lebiedzinska M, Duszynski J, Pinton P:Isolation of mitochondria-associated membranes and mitochondriafrom animal tissues and cells. Nat Protoc 2009, 4:1582e1590

29. Fernandez-Checa JC, Ookhtens M, Kaplowitz N: Effect of chronicethanol feeding on rat hepatocytic glutathione. Compartmentation,efflux, and response to incubation with ethanol. J Clin Invest 1987,80:57e62

30. Tietze F: Enzymic method for quantitative determination of nano-gram amounts of total and oxidized glutathione: applications tomammalian blood and other tissues. Anal Biochem 1969, 27:502e522

31. Duncan IW, Culbreth PH, Burtis CA: Determination of free, total,and esterified cholesterol by high-performance liquid chromatog-raphy. J Chromatogr 1979, 162:281e292

32. Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP,Clark SG, Ron D: IRE1 couples endoplasmic reticulum load tosecretory capacity by processing the XBP-1 mRNA. Nature 2002,415:92e96

33. Engelking LJ, Kuriyama H, Hammer RE, Horton JD, Brown MS,Goldstein JL, Liang G: Overexpression of Insig-1 in the livers oftransgenic mice inhibits SREBP processing and reduces insulin-stimulated lipogenesis. J Clin Invest 2004, 113:1168e1175

34. Canepa E, Borghi R, Vina J, Traverso N, Gambini J, Domenicotti C,Marinari UM, Poli G, Pronzato MA, Ricciarelli R: Cholesterol andamyloid-beta: evidence for a cross-talk between astrocytes andneuronal cells. J Alzheimers Dis 2011, 25:645e653

35. Wolf A, Bauer B, Hartz AM: ABC transporters and the Alzheimer’sdisease enigma. Front Psychiatry 2012, 3:54

36. Akram A, Schmeidler J, Katsel P, Hof PR, Haroutunian V: Increasedexpression of cholesterol transporter ABCA1 is highly correlated withseverity of dementia in AD hippocampus. Brain Res 2010, 1318:167e177

The American Journal of Pathology - ajp.amjpathol.orgFLA 5.2.0 DTD � AJPA1729_proof

37. Kim WS, Bhatia S, Elliott DA, Agholme L, Kagedal K, McCann H,Halliday GM, Barnham KJ, Garner B: Increased ATP-bindingcassette transporter A1 expression in Alzheimer’s disease hippo-campal neurons. J Alzheimers Dis 2010, 21:193e205

38. Rone MB, Fan J, Papadopoulos V: Cholesterol transport in steroidbiosynthesis: role of protein-protein interactions and implications indisease states. Biochim Biophys Acta 2009, 1791:646e658

39. Hayashi T, Rizzuto R, Hajnoczky G, Su TP: MAM: more than just ahousekeeper. Trends Cell Biol 2009, 19:81e88

40. Hayashi T, Fujimoto M: Detergent-resistant microdomains determinethe localization of sigma-1 receptors to the endoplasmic reticulum-mitochondria junction. Mol Pharmacol 2010, 77:517e528

41. Marriott KS, Prasad M, Thapliyal V, Bose HS: Sigma-1 receptor atthe mitochondrial-associated endoplasmic reticulum membrane isresponsible for mitochondrial metabolic regulation. J Pharmacol ExpTher 2012, 343:578e586

42. Area-Gomez E, Del Carmen Lara Castillo M, Tambini MD, Guardia-Laguarta C, de Groof AJ, Madra M, Ikenouchi J, Umeda M, Bird TD,Sturley SL, Schon EA: Upregulated function of mitochondria-associated ER membranes in Alzheimer disease. EMBO J 2012, 31:4106e4123

43. Hedskog L, Pinho CM, Filadi R, Ronnback A, Hertwig L,Wiehager B, Larssen P, Gellhaar S, Sandebring A, Westerlund M,Graff C, Winblad B, Galter D, Behbahani H, Pizzo P, Glaser E,Ankarcrona M: Modulation of the endoplasmic reticulum-mitochondria interface in Alzheimer’s disease and related models.Proc Natl Acad Sci U S A 2013, 110:7916e7921

44. Simmen T, Aslan JE, Blagoveshchenskaya AD, Thomas L, Wan L,Xiang Y, Feliciangeli SF, Hung CH, Crump CM, Thomas G: PACS-2controls endoplasmic reticulum-mitochondria communication andBid-mediated apoptosis. EMBO J 2005, 24:717e729

45. de Brito OM, Scorrano L: Mitofusin 2 tethers endoplasmic reticulumto mitochondria. Nature 2008, 456:605e610

46. Campbell AM, Chan SH: The voltage dependent anion channel af-fects mitochondrial cholesterol distribution and function. Arch Bio-chem Biophys 2007, 466:203e210

47. Ye J, Rawson RB, Komuro R, Chen X, Dave UP, Prywes R,Brown MS, Goldstein JL: ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell2000, 6:1355e1364

48. Zeng L, Lu M, Mori K, Luo S, Lee AS, Zhu Y, Shyy JY: ATF6modulates SREBP2-mediated lipogenesis. EMBO J 2004, 23:950e958

49. Charman M, Kennedy BE, Osborne N, Karten B: MLN64 mediatesegress of cholesterol from endosomes to mitochondria in the absenceof functional Niemann-Pick Type C1 protein. J Lipid Res 2010, 51:1023e1034

50. Rosati F, Sturli N, Cungi MC, Morello M, Villanelli F, Bartolucci G,Finocchi C, Peri A, Serio M, Danza G: Gonadotropin-releasinghormone modulates cholesterol synthesis and steroidogenesis in SH-SY5Y cells. J Steroid Biochem Mol Biol 2011, 124:77e83

51. Mari M, Morales A, Colell A, Garcia-Ruiz C, Fernandez-Checa JC:Mitochondrial glutathione, a key survival antioxidant. AntioxidRedox Signal 2009, 11:2685e2700

52. Chan RB, Oliveira TG, Cortes EP, Honig LS, Duff KE, Small SA,Wenk MR, Shui G, Di Paolo G: Comparative lipidomic analysis ofmouse and human brain with Alzheimer disease. J Biol Chem 2012,287:2678e2688

53. Brown J 3rd, Theisler C, Silberman S, Magnuson D, Gottardi-Littell N, Lee JM, Yager D, Crowley J, Sambamurti K, Rahman MM,Reiss AB, Eckman CB, Wolozin B: Differential expression ofcholesterol hydroxylases in Alzheimer’s disease. J Biol Chem 2004,279:34674e34681

54. Heverin M, Bogdanovic N, Lutjohann D, Bayer T, Pikuleva I,Bretillon L, Diczfalusy U, Winblad B, Bjorkhem I: Changes in thelevels of cerebral and extracerebral sterols in the brain of patients withAlzheimer’s disease. J Lipid Res 2004, 45:186e193

15

1860

� 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 16: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Barbero-Camps et al

18611862186318641865186618671868186918701871187218731874187518761877187818791880188118821883188418851886188718881889189018911892189318941895189618971898189919001901190219031904190519061907190819091910191119121913191419151916191719181919192019211922

1923192419251926192719281929193019311932193319341935193619371938193919401941194219431944194519461947194819491950195119521953195419551956195719581959196019611962196319641965

55. Bosch M, Mari M, Herms A, Fernandez A, Fajardo A, Kassan A,Giralt A, Colell A, Balgoma D, Barbero E, Gonzalez-Moreno E,Matias N, Tebar F, Balsinde J, Camps M, Enrich C, Gross SP, Garcia-Ruiz C, Perez-Navarro E, Fernandez-Checa JC, Pol A: Caveolin-1deficiency causes cholesterol-dependent mitochondrial dysfunctionand apoptotic susceptibility. Curr Biol 2011, 21:681e686

56. Webber KM, Stocco DM, Casadesus G, Bowen RL, Atwood CS,Previll LA, Harris PL, Zhu X, Perry G, Smith MA: Steroidogenicacute regulatory protein (StAR): evidence of gonadotropin-inducedsteroidogenesis in Alzheimer disease. Mol Neurodegener 2006, 1:14

57. Kil IS, Lee SK, Ryu KW, Woo HA, Hu MC, Bae SH, Rhee SG:Feedback control of adrenal steroidogenesis via H2O2-dependent,reversible inactivation of peroxiredoxin III in mitochondria. Mol Cell2012, 46:584e594

58. Caron KM, Soo SC, Wetsel WC, Stocco DM, Clark BJ, Parker KL:Targeted disruption of the mouse gene encoding steroidogenic acuteregulatory protein provides insights into congenital lipoid adrenalhyperplasia. Proc Natl Acad Sci U S A 1997, 94:11540e11545

59. Sano R, Annunziata I, Patterson A, Moshiach S, Gomero E,Opferman J, Forte M, d’Azzo A: GM1-ganglioside accumulation atthe mitochondria-associated ER membranes links ER stress toCa(2þ)-dependent mitochondrial apoptosis. Mol Cell 2009, 36:500e511

60. Shioda N, Ishikawa K, Tagashira H, Ishizuka T, Yawo H,Fukunaga K: Expression of a truncated form of the endoplasmicreticulum chaperone protein, sigma1 receptor, promotes mitochon-drial energy depletion and apoptosis. J Biol Chem 2012, 287:23318e23331

61. Rodriguez-Agudo D, Ren S, Wong E, Marques D, Redford K, Gil G,Hylemon P, Pandak WM: Intracellular cholesterol transporter StarD4binds free cholesterol and increases cholesteryl ester formation.J Lipid Res 2008, 49:1409e1419

62. Korytowski W, Rodriguez-Agudo D, Pilat A, Girotti AW: StarD4-mediated translocation of 7-hydroperoxycholesterol to isolatedmitochondria: deleterious effects and implications for steroidogenesisunder oxidative stress conditions. Biochem Biophys Res Commun2010, 392:58e62

63. Soccio RE, Adams RM, Maxwell KN, Breslow JL: Differential generegulation of StarD4 and StarD5 cholesterol transfer proteins. Acti-vation of StarD4 by sterol regulatory element-binding protein-2 andStarD5 by endoplasmic reticulum stress. J Biol Chem 2005, 280:19410e19418

64. Rodriguez-Agudo D, Ren S, Hylemon PB, Redford K, Natarajan R,Del Castillo A, Gil G, Pandak WM: Human StarD5, a cytosolicStAR-related lipid binding protein. J Lipid Res 2005, 46:1615e1623

65. Rodriguez-Agudo D, Calderon-Dominguez M, Medina MA, Ren S,Gil G, Pandak WM: ER stress increases StarD5 expression by

16FLA 5.2.0 DTD � AJPA1729_proo

stabilizing its mRNA and leads to relocalization of its protein fromthe nucleus to the membranes. J Lipid Res 2012, 53:2708e2715

66. Zheng Z, Zhang C, Zhang K: Role of unfolded protein response inlipogenesis. World J Hepatol 2010, 2:203e207

67. Fernandez A, Matias N, Fucho R, Ribas V, Von Montfort C, Nuno N,Baulies A, Martinez L, Tarrats N, Mari M, Colell A, Morales A,Dubuquoy L, Mathurin P, Bataller R, Caballeria J, Elena M,Balsinde J, Kaplowitz N, Garcia-Ruiz C, Fernandez-Checa JC:ASMase is required for chronic alcohol induced hepatic endoplasmicreticulum stress and mitochondrial cholesterol loading. J Hepatol2013, 59:805e813

68. Mari M, Morales A, Colell A, Garcia-Ruiz C, Kaplowitz N, Fer-nandez-Checa JC: Mitochondrial glutathione: features, regulation androle in disease. Biochim Biophys Acta 2013, 1830:3317e3328

69. Katayama T, Imaizumi K, Sato N, Miyoshi K, Kudo T, Hitomi J,Morihara T, Yoneda T, Gomi F, Mori Y, Nakano Y, Takeda J,Tsuda T, Itoyama Y, Murayama O, Takashima A, St George-Hyslop P, Takeda M, Tohyama M: Presenilin-1 mutations down-regulate the signalling pathway of the unfolded-protein response. NatCell Biol 1999, 1:479e485

70. Ricobaraza A, Cuadrado-Tejedor M, Marco S, Perez-Otano I, Garcia-Osta A: Phenylbutyrate rescues dendritic spine loss associated withmemory deficits in a mouse model of Alzheimer disease. Hippo-campus 2012, 22:1040e1050

71. Ricobaraza A, Cuadrado-Tejedor M, Perez-Mediavilla A, Frechilla D,Del Rio J, Garcia-Osta A: Phenylbutyrate ameliorates cognitivedeficit and reduces tau pathology in an Alzheimer’s disease mousemodel. Neuropsychopharmacology 2009, 34:1721e1732

72. Wiley JC, Pettan-Brewer C, Ladiges WC: Phenylbutyric acid reducesamyloid plaques and rescues cognitive behavior in AD transgenicmice. Aging Cell 2011, 10:418e428

73. Shafaati M, O’Driscoll R, Bjorkhem I, Meaney S: Transcriptionalregulation of cholesterol 24-hydroxylase by histone deacetylase in-hibitors. Biochem Biophys Res Commun 2009, 378:689e694

74. Pipalia NH, Cosner CC, Huang A, Chatterjee A, Bourbon P,Farley N, Helquist P, Wiest O, Maxfield FR: Histone deacetylaseinhibitor treatment dramatically reduces cholesterol accumulation inNiemann-Pick type C1 mutant human fibroblasts. Proc Natl Acad SciU S A 2011, 108:5620e5625

75. Kim SJ, Lee BH, Lee YS, Kang KS: Defective cholesterol traffic andneuronal differentiation in neural stem cells of Niemann-Pick type Cdisease improved by valproic acid, a histone deacetylase inhibitor.Biochem Biophys Res Commun 2007, 360:593e599

76. Yoon SO, Park DJ, Ryu JC, Ozer HG, Tep C, Shin YJ, Lim TH,Pastorino L, Kunwar AJ, Walton JC, Nagahara AH, Lu KP,Nelson RJ, Tuszynski MH, Huang K: JNK3 perpetuates metabolicstress induced by Abeta peptides. Neuron 2012, 75:824e837

ajp.amjpathol.org - The American Journal of Pathology

1966196719681969197019711972197319741975197619771978197919801981198219831984

f � 19 May 2014 � 11:53 am � EO: AJP13_0447

Page 17: Endoplasmic Reticulum Stress Mediates Amyloid β Neurotoxicity via Mitochondrial Cholesterol Trafficking

Q56

198519861987198819891990199119921993199419951996199719981999200020012002200320042005200620072008

200920102011201220132014201520162017201820192020202120222023202420252026202720282029203020312032

Supplemental Figure S1 Increased cholesterol levels and depleted mGSH content in 10-month-old APP/PS1 mice. Total cholesterol (A) and GSH (B)levels of brain homogenate and isolated mitochondria from WT and APP/PS1 transgenic mice at the indicated ages. **P < 0.01 versus WT mice values byunpaired two-tailed Student’s t-test (n Z 6 to 8). mth, month.

Supplemental Figure S2 Mitochondria isolated from brains of WT and APP/PS1 mice. After cellular subfraction, the purity of mitochondria wasconfirmed by determining in homogenate (Homo.), ER, or mitochondria (Mito.) the distribution of specific markers such as PERK and calreticulin (ER), Naþ/Kþ-ATPase a1 (plasma membrane), Rab5a (early endosomes), Rab7 (late endosomes/lysosomes), and cyclooxygenase 4 (COX-IV; mitochondria) using immunoblotanalysis. The densitometric values of the bands representing the specific antibody immunoreactivity were expressed as relative intensity compared withhomogenate values (n Z 3).

Supplemental Figure S3 Stable expression levels of cholesterol-related proteins in WT mice. Representative immunoblots of SREBP-2 and HMG-CoAreductase (HMGCR) (A) and the cholsterol carriers StAR and StARD3 (B) in brain extracts from WT and APP/PS1 mice at the indicated ages. The densitometricvalues of the bands representing the specific antibody immunoreactivity were normalized to the values of the corresponding b-actin band and expressed asrelative intensity compared with 4-month-old WT mice values. **P < 0.01 versus 4-month-old WT mice values by one-way analysis of variance with theDunnett post hoc test (n Z 3). mth, month.

Supplemental Figure S4 Undetectable signs of UPR response in WT mice. A and B: Representative immunoblots showing the expression of thechaperonic protein GRP78 and the ER stress signaling pathway proteins PERK, phospho-PERK, eIF2a, phospho-eIF2a, and CHOP in brain extracts from WT andAPP/PS1 (A/P) mice at the indicated ages. The densitometric values of the bands representing the specific antibody immunoreactivity were normalized to thevalues of the corresponding b-actin band and expressed as relative intensity compared with WT mice values. *P < 0.05 versus 4-month-old mice values;yP < 0.05 versus WT mice values by one-way analysis of variance with the Dunnett post hoc test (n Z 3). mth, month.

Supplemental Figure S5 Age-dependent Ab accumulation in APP/PS1 mice. Representative photomicrographs of hippocampus labeled with humanantieAb1-42 and counterstained with hematoxylin (n Z 3). Scale bar Z 100 mm.

FLA 5.2.0 DTD � AJPA1729_proof � 19 May 2014 � 11:53 am � EO: AJP13_0447