ebook img

Fig. 1 PDF

15 Pages·2015·3.22 MB·English
by  
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Fig. 1

TheJournalofNeuroscience,September2,2015•35(35):12137–12151•12137 NeurobiologyofDisease Neuronal-Targeted TFEB Accelerates Lysosomal (cid:2) Degradation of APP, Reducing A Generation and Amyloid Plaque Pathogenesis QingliXiao,1PingYan,1XiucuiMa,2,4HaiyanLiu,2,4RonaldoPerez,1AlecZhu,1ErnestoGonzales,1 XDanielleL.Tripoli,1XLeahCzerniewski,1AndreaBallabio,5JohnR.Cirrito,1XAbhinavDiwan,2,3,4*and XJin-MooLee1* 1DepartmentofNeurologyandtheHopeCenterforNeurologicalDisorders,2CenterforCardiovascularResearchinDepartmentofMedicine,and 3DepartmentofCellBiologyandPhysiology,WashingtonUniversitySchoolofMedicine,St.Louis,Missouri63110,4JohnCochranVAMedicalCenter,St. Louis,Missouri63108,and5TelethonInstituteofGeneticsandMedicine,Pozzuoli,Naples,Italy80078 InAD,animbalancebetweenA(cid:2)productionandremovaldriveselevatedbrainA(cid:2)levelsandeventualamyloidplaquedeposition.APP undergoesnonamyloidogenicprocessingvia(cid:3)-cleavageattheplasmamembrane,amyloidogenic(cid:2)-and(cid:4)-cleavagewithinendosomesto generateA(cid:2),orlysosomaldegradationinneurons.Consideringmultiplereportsimplicatingimpairedlysosomefunctionasadriverof increasedamyloidogenicprocessingofAPP,weexploredtheefficacyoftargetingtranscriptionfactorEB(TFEB),amasterregulatorof lysosomalpathways,toreduceA(cid:2)levels.CMVpromoter-drivenTFEB,transducedviastereotactichippocampalinjectionsofadeno- associatedvirusparticlesinAPP/PS1mice,localizedprimarilytoneuronalnucleiandupregulatedlysosomebiogenesis.Thisresultedin reductionofAPPprotein,the(cid:3)and(cid:2)C-terminalAPPfragments(CTFs),andinthesteady-stateA(cid:2)levelsinthebraininterstitialfluid. Inagedmice,totalA(cid:2)levelsandamyloidplaqueloadwereselectivelyreducedintheTFEB-transducedhippocampi.TFEBtransfectionin N2acellsstablyexpressingAPP695,stimulatedlysosomebiogenesis,reducedsteady-statelevelsofAPPand(cid:3)-and(cid:2)-CTFs,andatten- uatedA(cid:2)generationbyacceleratingfluxthroughtheendosome-lysosomepathway.Cycloheximidechaseassaysrevealedashorteningof APPhalf-lifewithexogenousTFEBexpression,whichwaspreventedbyconcomitantinhibitionoflysosomalacidification.Thesedata indicate that TFEB enhances flux through lysosomal degradative pathways to induce APP degradation and reduce A(cid:2)generation. ActivationofTFEBinneuronsisaneffectivestrategytoattenuateA(cid:2)generationandattenuateamyloidplaquedepositioninAD. Keywords: Alzheimer’sdisease;amyloid;amyloidprecursorprotein;lysosome;TFEB SignificanceStatement AkeydriverforADpathogenesisisthenetbalancebetweenproductionandclearanceofA(cid:2),themajorcomponentofamyloid plaques.Herewedemonstratethatlysosomaldegradationofholo-APPinfluencesA(cid:2)productionbylimitingtheavailabilityof APPforamyloidogenicprocessing.UsingviralgenetransferoftranscriptionfactorEB(TFEB),amasterregulatoroflysosome biogenesisinneuronsofAPP/PS1mice,steady-statelevelsofAPPwerereduced,resultingindecreasedinterstitialfluidA(cid:2)levels andattenuatedamyloiddeposits.TheseeffectswerecausedbyacceleratedlysosomaldegradationofendocytosedAPP,reflected byreducedAPPhalf-lifeandsteady-statelevelsinTFEB-expressingcells,withresultantdecreaseinA(cid:2)productionandrelease. Additionalstudiesareneededtoexplorethetherapeuticpotentialofthisapproach. Introduction InAD,increasedgenerationoftheA(cid:2)peptideinneuronssyner- gizeswithimpairedA(cid:2)removalfromtheinterstitialfluid(ISF)in toJ.R.C.WethankWimAnnaert,VIB(FlandersInstituteforBiotechnology),Leuven,Belgium,forprovidingthedualfluorescentAPP constructandStuartKornfeld,WashingtonUniversity,forscientificinput. Theauthorsdeclarenocompetingfinancialinterests. *A.D.andJ.-M.L.contributedequallytothiswork. ReceivedFeb.19,2015;revisedJuly2,2015;acceptedJuly24,2015. Correspondenceshouldbeaddressedtoeitherofthefollowing:Jin-MooLee,ProfessorofNeurology,660South Authorcontributions:Q.X.,J.R.C.,A.D.,andJ.-M.L.designedresearch;Q.X.,P.Y.,X.M.,H.L.,R.P.,A.Z.,E.G.,D.L.T., EuclidAvenue,CB8111,St.Louis,MO63110.E-mail:[email protected];orAbhinavDiwan,AssistantProfessorof L.C.,J.R.C.,andA.D.performedresearch;A.B.contributedunpublishedreagents/analytictools;Q.X.,P.Y.,X.M.,H.L., Medicine, Cell Biology and Physiology, 4940 Parkview Place, CSRB 827, St. Louis, MO 63110. E-mail: A.Z.,D.L.T.,L.C.,J.R.C.,A.D.,andJ.-M.L.analyzeddata;Q.X.,J.R.C.,A.D.,andJ.-M.L.wrotethepaper. [email protected]. ThisstudywassupportedbygrantsfromtheAlzheimer’sAssociation(NIRG12-242588)toA.D.,BrightfocusFoundation DOI:10.1523/JNEUROSCI.0705-15.2015 (A2012151)toJ.R.C.,andfromtheNationalInstitutesofHealth(R21NS082529)toJ.-M.Land(R01AG042513andR21AG045691) Copyright©2015theauthors 0270-6474/15/3512137-15$15.00/0 12138•J.Neurosci.,September2,2015•35(35):12137–12151 Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation driving extracellular amyloid plaque formation (Haass et al., milliliterintobilateralhippocampiat6monthsofage,asdescribedpre- 2012)andtheresultantneuropathogenesis(Bloom,2014).After viously(Xiaoetal.,2012).Wild-typemiceofeithersexwerealsoinjected synthesisintheendoplasmicreticulum,APPistraffickedfrom withAAV8-CMV-FLAG-TFEBorAAV8-CMV-GFPat4monthsofage the Golgi to the plasma membrane, where it is cleaved by toassesstheeffectofTFEBtransductiononendogenousAPPprocessing (cid:3)-secretaseswithintheA(cid:2)peptidefragmentregion,inamanner after4months.Beforeinjection,AAVparticlesweresubjectedtotwo thatprecludesA(cid:2)generation(i.e.,nonamyloidogenicor(cid:3)-cleav- roundsofcesiumchloridegradientpurificationandtiterswerequanti- fiedbyassessmentofviralgenomes.Forplaqueloadstudies,TFEBor age;RajendranandAnnaert,2012).Alternately,APPisendocy- control-transduced APP/PS1 mice (six males and eight females per tosed, and sequentially cleaved by a (cid:2)-secretase (BACE1) and group) were killed 4 months later. One hemisphere was fixed and then the (cid:4)-secretase complex (comprised of presenilins, nicas- coronal sections (50 (cid:5)m) were cut for histological analysis (X-34 trin,APH-1,andPEN2)withinendosomes,togenerateA(cid:2)pep- stainingandHJ3.4immunostaining).Theotherhemispherewassnap tides (amyloidogenic cleavage; Rajendran and Annaert, 2012). frozen on dry ice and stored at (cid:4)80°C for biochemical analysis Accumulatingevidencesuggeststhatdysregulationofendocyto- (ELISAandimmunoblotting).ToassesstheabundanceofA(cid:2)species sis(Haroldetal.,2009;Ginsbergetal.,2010)withaccumulation inpredepositingmice,theabovementionedAAVviralvectorswere of large endosomes within neurons, as observed in preclinical injected into the hippocampi of APP/PS1 mice of either sex at 2 monthsofageandtheanimalswerekilled1monthlater.Allanimal stages of AD pathology (Cataldo et al., 2000) and in neurons care and surgical procedures were approved by the Animal Studies transdifferentiatedfromfibroblastsofpatientswithAD(Israelet CommitteeofWashingtonUniversitySchoolofMedicineinaccor- al.,2012),increasestheproclivityforamyloidogenicprocessing dancewithguidelinesoftheNIH. ofAPP. In vivo microdialysis. AAV particles driving expression of TFEB EndocytosedAPPcanbesortedbacktotheGolgi(Morelet (AAV8-CMV-FLAG-TFEB) or GFP as control (AAV8-CMV-GFP) al., 2013) or trafficked to lysosomes along with other endo- weretransducedbystereotacticallyguidedinjectioninthehippocam- somal cargo (The´ry et al., 2002). Indeed, a large fraction of pusof2-month-oldAPP/PS1transgenicmiceofeithersexfollowed full-lengthAPPisdegradedwithinlysosomes(Caporasoetal., byinvivomicrodialysis,1monthlater,aspreviouslydescribed(Cir- 1992,1994;Goldeetal.,1992;Haassetal.,1992).Experimen- ritoetal.,2003,2011).Allstudieswereinitiatedatthesametimeof tal inhibition of lysosome function markedly prolongs APP day. Briefly, a guide cannula (BR-style; Bioanalytical Systems) was half-life(Caporasoetal.,1992)andimpairsendocytoticflux implanted and cemented with the tip at coordinates: bregma (cid:4)3.1 mm,midline(cid:4)2.5mm,1.2mmbelowduraata12°angle.A2mm withaccumulationofenlargedvesiclesindystrophicneurites microdialysis probe was then inserted into the hippocampus that mimicking AD pathology (Lee et al., 2011), with increased containeda38kDaMWCOsemipermeablemembrane(Bioanalytical BACE1(Chiaetal.,2013)and(cid:2)-C-terminalfragment(Golde Systems)allowingmoleculessmallerthanthiscutofftodiffuseinto et al., 1992; Sannerud et al., 2011) resulting in markedly in- theprobe.A(cid:2)capableofenteringtheprobeisdubbed“exchangeable creasedA(cid:2)generation.Relevanttothisdiscussionisthatim- A(cid:2)” (eA(cid:2); Cirrito et al., 2003). The probe was flushed with 0.15% pairedlysosomalfunctionisobservedwithaging(Katoetal., bovineserumalbumin(Sigma)inanartificialCSFperfusionbufferat 1998;CuervoandDice,2000;Wolfeetal.,2013)andwithloss aconstantrate(1.0(cid:5)l/min).Theeffluatewascollectedintoarefrig- ofpresenilins(duetomutationscausingfamilialAD;Leeetal., eratedfractioncollectorandassayedbysandwichELISAforA(cid:2) x-40 2010; Neely et al., 2011; Coen et al., 2012), and is suspected peptidesattheendofeachexperiment.Duringmicrodialysis,animals withpolymorphismsingenescodingforlysosomalproteins, werehousedinspeciallydesignedcagestopermitfreemovementand adlibitumfoodandwaterwhileISFA(cid:2)wassampled.Baselinelevelsof whichconferincreasedriskforsporadicAD,includingphos- ISFA(cid:2)weresampledevery90minbetweenhours9and16(afterthe pholipase D3 (Cruchaga et al., 2014), cystatin C (Hua et al., microdialysisprobeisinserted)andaveragedtodeterminethe“base- 2012), and Cathepsin D (Schuur et al., 2011). Also, levels of lineISFA(cid:2)level”ineachmouse.Absoluteinvivoconcentrationof APP, its (cid:2)-CTF, and A(cid:2)are increased in mouse models of ISFeA(cid:2)foreachmousewasdeterminedbycorrectingforthe20.8% lysosome storage disorders, namely, Neimann-Pick disease recovery (1.0 (cid:5)l/min) as obtained by the interpolated zero flow (Kodametal.,2010),mucolipidoses(Keilanietal.,2012),and methoddescribedpreviously(Menacherryetal.,1992;Cirritoetal., sphingolipidoses (Tamboli et al., 2011). These observations 2003).Athour16(t(cid:5)0),a(cid:4)-secretaseinhibitor,CompoundE(200 incriminate lysosomal dysfunction as the common cellular nM reverse microdialysis; synthesized by AsisChem), was adminis- mechanism in provoking increased A(cid:2)generation and im- tereddirectlytothehippocampusbyaddingthedrugtothemicrodi- pairedA(cid:2)removal(FunkandKuret,2012;PericandAnnaert, alysisperfusionbuffer.ISFA(cid:2)wasthensampledevery60minforan 2015). additional8h.Thisenabledmeasurementoftheeliminationhalf-life ofendogenousISFA(cid:2)invivoasdescribedpreviously(Cirritoetal., Asatherapeuticstrategytoaddresslysosomalinsufficiencyin 2003). astrocytes, we previously evaluated activation of transcription Immunohistochemistry.Frozenbrainsectionswereincubatedin0.3% factor EB (TFEB), a master inducer of lysosomal degradative H O inTBSfor10minandblockedwith3%drymilkinTBS-X(0.25% pathways(Settembreetal.,2013a),anddemonstratedthatastro- 2 2 TritonX-100inTBS)for1h,followedbyincubationwithHJ3.4antibody cytic TFEB expression stimulates A(cid:2)uptake and its lysosomal (anti-A(cid:2)-1-13;Rohetal.,2012)overnight.ThereaftersolutionAfrom degradationtoreduceISFA(cid:2)(Xiaoetal.,2014).Inthisstudy,we VectastainABCkit(1:400)wasappliedfor1h,followedby0.025%DAB evaluatedtheefficacyofneuronalTFEBactivationinenhancing in0.25%NiCl and0.05%H O for10–15min.Thesliceswereplaced 2 2 2 lysosomaldegradationofAPPtoreduceA(cid:2)levelsandattenuate onglassslides,driedovernight,dehydratedandmounted,andimaged amyloidplaquepathogenesis. withconfocalmicroscopy(ZeissLSM). X-34plaquestaining.BrainslicesweremountedonSuperFrostPlus slides,permeabilizedwith0.25%TritonX-100for30min,andstained MaterialsandMethods withX-34(dissolvedin40%ethanol,60%water,pH10.0;agenerousgift Studiesinanimalmodels.APP/PS1transgenicmice(B6C3-Tg(APPswe/ fromRobertMach,WashingtonUniversity,St.Louis,MO)for20min. PS1(cid:2)E9)85Dbo/Mmjax)ofeithersexwereobtainedfromTheJackson ThereafterthetissuewasthoroughlyrinsedinPBSandmountedwith Laboratory(Jankowskyetal.,2004)andinjectedwithAAV8viralparti- Fluoromountmountingmedia. cles(generatedbytheHopeCenterViralCoreatWashingtonUniversity) Amyloidplaquequantification.Fiftymicrometerthickbrainsections drivingexpressionofTFEB(AAV8-CMV-FLAG-TFEB)orGFPascon- werecollectedevery300(cid:5)msfromtherostralanteriorcommissureto trol(AAV8-CMV-GFP),eachwith2(cid:5)lof1.5(cid:3)1012viralgenomesper caudal hippocampus. For plaque imaging, sections were stained with Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation J.Neurosci.,September2,2015•35(35):12137–12151•12139 X-34orimmunostainedwithHJ3.4anti-A(cid:2)antibodies.High-resolution agedmice(10monthsold,whenplaquesareabundant),hippocampal digitalimagesofthestainedbrainsliceswereobtainedwiththeNanoZo- tissuesweresequentiallyhomogenizedinPBSfollowedby5Mguanidine omerDigitalScanner(HamamatsuPhotonics).Thetotalareaofplaque in TBS, pH 8.0 (to extract fibrillar and membrane-bound A(cid:2)). For coveragewasmeasuredusingNIHImageJintheregionofthehippocam- ELISA assays, A(cid:2) and A(cid:2) peptides were captured with mouse x-40 x-42 pusorpiriformcortex(devoidofviraltransduction)andexpressedas monoclonal-coating antibodies HJ2 (anti-A(cid:2)35-40) and HJ7.4 (anti- percentagetotalareaforeachslice.Resultsfromn(cid:5)4sectionswere A(cid:2)37-42)orHJ5.1(anti-A(cid:2)13-28),respectively,aspreviouslydescribed averagedtorepresenteachanimal. (Kimetal.,2009;Xiaoetal.,2014).HJ5.1(anti-A(cid:2)13-28),abiotinylated Immunofluorescence.Paraffin-embeddedbrainsectionswerewashed antibodytargetingthecentraldomain,orHJ3.5(anti-A(cid:2)1-13,provided withPBSandpermeabilizedwith0.3%TritonX-100inPBSfor20min byDr.DavidHoltzman,WashingtonUniversitySchoolofMedicine,St. followedbyblocking.Fordoublelabeling,amixtureofthefollowing Louis,MO),whichtargetstheN-terminalaminoacids,wasusedasthe antibodies was used: mouse anti-TFEB (MyBioSource, 1:100), rabbit detecting antibody, followed by streptavidin-poly-HRP-40 (Fitzgerald anti-NeuN(Abcam;1:100),rabbitanti-GFAP(Sigma;1:100),andrab- Industries).AllELISAassaysweredevelopedusingSuperSlowELISA bitanti-Iba1(WakoChemicals;1:1000).Asecondaryantibodymixture TMB(Sigma)andabsorbancereadonaBio-TekEpochplatereaderat of Alexa Fluor 488-conjugated goat anti-mouse IgG and Alexa Fluor 650nm.StandardcurvesweregeneratedfromsynthetichumanA(cid:2) 1–40 594-conjugatedgoatanti-rabbitIgG(Invitrogen)wasapplied.Sections orA(cid:2) peptides(AmericanPeptide). 1–42 wereimagedbyconfocalmicroscopy(ZeissLSM). QuantitativePCR.Real-timePCRwasperformedasdescribedprevi- Neuronalcounts.Stereologicalanalysisforneuronalcountswasper- ously(Maetal.,2012).Briefly,totalRNAwaspreparedfromtransfected formedonsectionsstainedwith0.05%cresylvioletsolution,asprevi- N2a-APP695cellsorAAV-transducedhippocampususinganRNA-easy ouslydescribed(Xiaoetal.,2014).Briefly,cresylviolet-stainedcellswith minikit(Qiagen),andcDNAwassynthesizedwith1(cid:5)goftotalRNA nuclei imaged within the inclusive zone of each dissector frame were usingtheSuperScriptIIIfirst-strandsynthesissystem(Invitrogen).One counted in four equally spaced sections (50 (cid:5)m apart) in the dorsal microliterofcDNAtemplatewasmixedwith12.5(cid:5)lof2(cid:3)SYBRGreen hippocampus of strata pyramidale CA1 and CA3, using a computer- PCRMasterMix(Invitrogen)andsubjectedtoquantitativePCRintrip- basedstereologysystem(StereoInvestigator;MicroBrightField)withthe licateunderthefollowingconditions:50°C,2min;95°C,10min;fol- opticalfractionator(Westetal.,1991). lowed by 40 cycles of 95°C, 15 s; 60°C, 1 min in the ABI7500 Fast In vitro studies. N2a cells stably expressing the human APP695 RealTimePCRsystem.ThehousekeepinggeneGAPDHwasalsoampli- construct (N2a-APP695) were grown in DMEM/Opti-MEM (50:50) fiedinparallelasareferenceforthequantificationoftranscripts.Primer supplementedwith5%FBSand200(cid:5)g/mlG418andtransfectedwith sequencesusedhavebeenpreviouslydescribed(Xiaoetal.,2014). FLAG-tagged murine TFEB or empty vector, as described previously Immunoblotting.Proteinsampleswererunon4–12%Bis-Trisgels. (Xiao et al., 2014). We observed (cid:6)90% efficiency for transfection of Blotswereprobedwiththefollowingantibodies:FLAG(Sigma;1:1000), exogenousTFEB(orGFPascontrol)intheseinvitrostudies.Forassess- TFEB (MyBioSource; 1:500), LAMP1 (Santa Cruz Biotechnology; mentofAPPhalf-lifebycycloheximidechaseassay,N2a-APP695cells 1:500),CathepsinB(Abcam;1:500),CathepsinD(agenerousgiftfrom transfectedwithTFEB(orcontrol)weretreatedwith50(cid:5)g/mlcyclohex- StuartKornfeld,WashingtonUniversity,St.Louis,MO;1:2000),APP imidefollowedbypreparationofcellularextractsatvarioustimeinter- CT695(todetectbothfull-lengthAPPandC-terminalfragments;Invit- valsforimmunoblottingforAPP.Toinhibitlysosomeacidificationand rogen;1:500),6E10(todetectsAPP(cid:3);Covance;1:1000),Adam10(Santa function,studieswereperformedinthepresenceofbafilomycinA1(100 CruzBiotechnology;1:100),Adam17(Millipore;1:1000),BACE1(Cell nM),aspreviouslydescribed(Xiaoetal.,2014).Studieswithcell-surface Signaling Technology; 1:1000), Psen1 (Santa Cruz Biotechnology; biotinylation to assess internalization of APP were performed as de- 1:200),nicastrin(ThermoScientific;1:1000),IDE(Abcam;1:2000),ne- scribed previously (Xiao et al., 2012). Briefly, cells cultured in 6-well prilysin (Millipore; 1:500), MMP2 (Abcam; 1:1000), MMP9 (Abcam; plateswerewashedwithcoldPBSandsurfaceproteinswerelabeledwith 1:1000), HRP-conjugated biotin (Cell Signaling Technology; 1:1000), the nonmembrane-permeant, cleavable biotin derivative, sulfo-NHS- and actin (Sigma; 1:2000). Normalized band intensity was quantified SS-biotin(1mg/mlinPBS).Cellswerekeptat4°Cfor30mininthedark usingImageJsoftware. andgentlyrockedduringtheincubationperiod.Thebiotinreagentwas Statistical analysis. All data are shown as Mean (cid:7) SEM. Data were quenchedbytreatingthecellswithtwo15minwashesof0.1Mglycinein analyzedbytwo-tailedStudent’sttestfordetectingsignificantdiffer- PBS.CellswererinsedwithPBSandlysedinRIPAbuffercontaining50 encesbetweentwogroups.Differencesamongmultiplegroupswerean- mMTris-HCl,pH7.5;150mMNaCl;1%(v/v)NonidetP-40;0.5%(w/v) alyzedbyone-wayandtwo-wayANOVAforoneandtwoindependent deoxycholate; and 1(cid:3) protease inhibitor mixture. Lysates were incu- variables,respectively,followedbyposthocTukey’stest.Statisticalsignif- batedwithDynabeadsMyOneStreptavidinT1(Invitrogen)for2hina icancewaspeggedatp(cid:8)0.05(*p(cid:8)0.05and**p(cid:8)0.01). rotarymixertoisolatebiotin-labeledproteins.Acohortofsurfacebiotin- labeledcellswasincubatedfor0,1,2,3,5,7.5,10,20,and30minat37°Cto Results allowinternalization,followedbyrapidcoolingonicetostopendocytosis. AAV-CMV-TFEBtransducesTFEBspecificallyinneuronsof Tocleavebiotinexposedatthecellsurface,thesecellswereincubatedthree APP/PS1micewithupregulatedlysosomeabundance timesfor20minat4°Cwith50mM2-mercaptoethanesulfonicacid(Sigma) Lysosomaldysfunctionisspeculatedtoplayacentralroleinen- in50mMTris-HCl,pH8.7;100mMNaCl;and2.5mMCaCl2.Afterthorough hancedA(cid:2)generationinneurons(Leeetal.,2010;Neelyetal., rinsingwithPBScontaining20mMHEPES,cellswerelysedinRIPAbuffer, 2011; Coen et al., 2012; Funk and Kuret, 2012; McBrayer and and internalized biotinylated proteins were immunoprecipitated with streptavidinfollowedbyimmunoblottingforAPP(CT695;Invitrogen).Up- Nixon,2013;Wolfeetal.,2013)oritsimpairedclearance(Wyss- takeofbiotin-labeledtransferrin(T3915;Sigma)wasassessedinparallelin Corayetal.,2003;Majumdaretal.,2011;Leeetal.,2012;Kane- TFEB and vector-transfected cells to determine the effect of TFEB on kiyo et al., 2013). In previous studies, we demonstrated that clathrin-mediated endocytosis. Immunofluorescence was performed on targetingexpressionofTFEB,amasterinduceroflysosomalbio- C17.2cellscotransfectedwithCer-APP-YFPandTFEBorcontrolvectorand genesis(Sardielloetal.,2009;Settembreetal.,2011,2013b),spe- incubatedwithLysoTrackerRed(L7526;Invitrogen)tolabelthelysosome cifically to astrocytes, stimulated lysosome biogenesis and compartment,andlivecellswereexaminedbyconfocalmicroscope(Zeiss function resulting in accelerated A(cid:2)uptake and degradation LSM). (Xiaoetal.,2014).Inthecurrentstudy,weexploredthehypoth- A(cid:2)ELISA.A(cid:2)speciesincelllysatesandmediafromtransfectedN2a- esisthatenhancinglysosomalbiogenesisinneuronswithexoge- APP695weredetectedbysandwichELISA,aspreviouslydescribed(Xiao et al., 2014). To detect total A(cid:2)in the hippocampus in 3-month-old nousexpressionofTFEBwillstimulatelysosomaldegradationof mice,dissectedtissuewassequentiallyhomogenizedinPBSfollowedby holo-APP to attenuate A(cid:2)production. Accordingly, we per- RIPAbuffertoobtaindetergent-solubleA(cid:2)atanagewhenplaquesare formedstereotacticallyguidedinjectionsofAAV8particlesen- notobserved(Yanetal.,2009),andsampleswerepooledforanalysis.In coding a CMV promoter-driven construct to transduce TFEB 12140•J.Neurosci.,September2,2015•35(35):12137–12151 Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation (AAV8-CMV-FLAG-TFEB; and AAV8- CMV-GFP as control) in the hippocam- pus of 6-month-old APP/PS1 mice, at a stage of early plaque deposition and growth (Yan et al., 2009). Stereotactic AAV injections consistently resulted in widespreadtransductioninthehippocam- pus(datanotshown).Asweobservedpre- viously (Xiao et al., 2012), the CMV promoter-driven construct specifically re- sultedinTFEBexpressioninneuronswitha neuron-specificmarker,NeuN,andnot inastrocytesormicroglia(Fig.1A).Im- portantly,exogenousTFEBlocalizedto the neuronal nuclei (Fig. 1A) and hip- pocampal extracts from AAV8-TFEB- transduced mice demonstrate robust overexpression with TFEB (11-fold in- creaseovercontrol,p(cid:8)0.01;N(cid:5)3per group)withincreasedabundanceoflys- osomal proteins, LAMP1, and Cathep- sinBandCathepsinD(Fig.1B,C),two lysosomalproteasesthathavebeenim- plicated in APP degradation (Higaki et al.,1996;Mueller-Steineretal.,2006). NeuronalTFEBexpressiondrives reducedAPPabundance,invivo APP695issynthesizedandprocessedpri- marily in neurons, where its sequential cleavageviathe(cid:2)-and(cid:4)-secretasesresults in generation of amyloidogenic A(cid:2)pep- tides(Hartmannetal.,1997;Kamenetzet al.,2003).Alternately,APPprocessingcan proceedvia(cid:3)-secretasesinamannerthat Figure1. AAV8-CMV-TFEBdriveslysosomebiogenesisinneurons.A,Representativeconfocalimagesdemonstratingexpres- precludes A(cid:2) generation (Haass et al., sionofTFEB(green)invariousCNScelltypesmarkedbytheexpressionofNeuN(neurons;red,top),GFAP(astrocytes;red,middle), andIba-1(microglia;red,bottom)inthehippocampusofAPP/PS1miceinjectedwithAAV8-CMV-FLAG-TFEBparticles.Theboxed 2012). Studies have demonstrated that a inserts(upperleftcorners)demonstratemagnifiedimagesofTFEB-expressingcells.B,C,Immunoblots(B)withquantification(C) largefractionoffull-lengthAPPproteinis oflysosomalproteinsinextractsfromhippocampitransducedwithAAV8-CMV-FLAG-TFEB(TFEB)orAAV8-CMV-GFP(GFP).N(cid:5)3 also degraded within lysosomes, which pergroup;**p(cid:8)0.01. maylimitthepoolofAPPavailablefor(cid:3)-, (cid:2)-,and(cid:4)-cleavage(Caporasoetal.,1992, thesedatasuggestthatTFEBexpressionfacilitatesholo-APPdegra- 1994;Goldeetal.,1992).GiventhatTFEBdrivesupregulationof dation via pathways that do not involve APP processing by the lysosomal machinery, we examined the effect of neuronal (cid:3)-cleavageorsequential(cid:2)-and(cid:4)-cleavage. TFEBexpressiononabundanceofAPPanditsvariouscleavage fragments. TFEB-transduced hippocampal extracts demon- strateda28%reductioninAPPproteinabundance,accompanied NeuronalTFEBexpressionreducesISFA(cid:2)levels,invivo byamatching31and36%reductionin(cid:3)-CTFand(cid:2)-CTF(Fig. A(cid:2)peptides are released into the ISF and removed via cellular 2A,B).Givenpreviousobservationsthatamyloidpathogenesisis uptake and degradation, extracellular proteolysis, or transport acceleratedinfemalemice(Callahanetal.,2001),weexamined across the blood–brain barrier. Consequently, the steady-state theeffectsofTFEBtransductioninmaleandfemalemicesepa- levelofA(cid:2)intheISFisdeterminedbythebalancebetweenitsrate rately. We observed comparable reductions in APP in the two of production and degradation. Considering that exogenous sexes(by29%inmalesvs30%infemales;datanotshown).These TFEBexpressionresultsinareductionintotalAPPlevelswitha datashowthattheeffectofneuronalTFEBexpressiononAPP parallel reduction in abundance of its cleaved fragments (Fig. abundanceisindependentofgender. 2A,B), we hypothesized that TFEB transduction will attenuate ThereductioninAPPabundancewasnotcausedbyadifference A(cid:2)productionresultinginreducedA(cid:2)steady-statelevelsinthe in the transcript levels for the transgenically expressed human ISF. Accordingly, we transduced 2-month-old APP/PS1 mice APP695protein(Fig.2C)orintheexpressionof(cid:3)-secretasesand with AAV8-CMV-FLAG-TFEB and control AAV8-CMV-GFP componentsofthe(cid:2)-and(cid:4)-secretasemachinerybetweentheTFEB viralparticlesandperformedinvivomicrodialysis1monthlater and control-transduced hippocampi (Fig. 2C–E). Importantly, todeterminewhetherneuronalTFEBexpressionimpactsISFA(cid:2) TFEB-transducedhippocampididnotdemonstrateadifferencein atatimepointbeforedepositionofplaques(Yanetal.,2009)to neuronalcountscomparedwithcontrol(Fig.2F),rulingoutneuro- avoid a potential confounding influence from exchangeable naltoxicityastheunderlyingcausefortheseobservations.Together plaque-boundA(cid:2)pool.TFEB-transducedmicedemonstrateda withtheproportionatelycomparablereductionsinvariousCTFs, 57%reductioninsteady-stateISFA(cid:2)levelscomparedwithcon- Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation J.Neurosci.,September2,2015•35(35):12137–12151•12141 Figure2. NeuronalTFEBtransductionresultsinreducedAPPabundance.A,B,Immunoblot(A)withquantitation(B)ofAPPandits(cid:3)-and(cid:2)-CTFsinAPP/PS1mousehippocampitransducedwith AAV8-CMV-FLAG-TFEB(TFEB)orAAV8-CMV-GFP(GFP).N(cid:5)6pergroup(n(cid:5)3malesandn(cid:5)3females);**p(cid:8)0.01.Representativeimmunoblotisfrommalemousetissues.C,Expressionof transcriptscodingforvariouscomponentsoftheAPP-processingmachineryinhippocampaltissuestransducedwithAAV8-CMV-FLAG-TFEB(TFEB)orAAV8-CMV-GFP(GFP).N(cid:5)5pergroup.No statisticallysignificantdifferenceswerenotedforanyofthetranscripts.D,E,Immunoblot(D)andquantitation(E)ofAPP-processingmachineryproteinsinhippocampitransducedwithTFEB(orGFP ascontrol,asinA).N(cid:5)3pergroup.F,NeuronalcountsintheCA1andCA3layersofthehippocampifromAAV8-CMV-FLAG-TFEB(TFEB)andAAV8-CMV-GFP(GFP)-transducedAPP/PS1mice.N(cid:5) 4pergroup. trols(Fig.3A).Aftersteady-statemeasuresofISFA(cid:2)wereob- Importantly, total immunodetectable A(cid:2)levels were also tained, a potent (cid:4)-secretase inhibitor, Compound E, was reduced in the hippocampal extracts from TFEB-transduced administeredtoinhibitAPPcleavagetoA(cid:2)(Cirritoetal.,2011), mice at this early stage of amyloid pathogenesis, compared andISFwassampledforanadditionalperiodof8htodetermine with controls (Fig. 3F,G), confirming that the reduction in theeliminationrate(half-life)ofpre-existingA(cid:2)(Fig.3A),which steady-state levels of ISF A(cid:2)was not due to intracellular A(cid:2) was observed to follow first-order kinetics in both control and accumulation.TogetherwiththereductioninAPPabundance TFEB-transduced mice (Fig. 3B). We observed a modest 24% and that of its various cleaved fragments (Fig. 2A,B), these reductioninhalf-lifeofISFA(cid:2)inCMV-TFEB-transducedhip- datapointtoapredominanteffectofneuronalTFEBexpres- pocampi,indicatingthatneuronalTFEBtransductionalsoaccel- sion on attenuating A(cid:2)generation with a modest effect on eratesA(cid:2)removalfromISF(Fig.3B).Thatsteady-stateISFA(cid:2) accelerating A(cid:2) removal as the primary driver of reduced levelsarereducedby57%,buthalf-lifeisreducedbyonly24%, steady-stateISFA(cid:2)levels. stronglysuggeststhatTFEBhasonlyaminorroleinenhancing ISFA(cid:2)clearancewiththeprimaryeffectinsteadonreducingA(cid:2) NeuronalTFEBexpressionreducesA(cid:2)levelsandamyloid production. Indeed, previous studies have demonstrated that plaquesinAPP/PS1transgenicmice neuronstakeupA(cid:2)viaendocytosisandmacropinocytosis,andit ISFA(cid:2)levelscorrelatewithamyloidplaquepathology(Beroet isconceivablethatTFEBtransductionstimulatesA(cid:2)uptakein al.,2011),supportingthehypothesisthatA(cid:2)aggregatesintoam- neurons via the latter similar to our studies with astrocyte- yloidplaquesinaconcentration-dependentmanner,invivo.In- targetedTFEBexpression(Xiaoetal.,2014).Importantly,the deed, studies from our laboratory as well as others have effectofexogenousTFEBonreductioninISFA(cid:2)levelsandA(cid:2) demonstratedthatstrategiestoreduceISFA(cid:2)levelsdriveare- half-lifewascomparableinbothsexes(datanotshown).We ductioninamyloidplaquedeposition(Crameretal.,2012;She- also explored the possibility that TFEB transduction could lineetal.,2014;Xiaoetal.,2014).Accordingly,wepositedthat enhanceextracellulardegradationofA(cid:2),butdidnotobserve neuronalTFEBtransductionwilldrivereductioninfibrillarA(cid:2) any alteration in the transcript levels for known A(cid:2)- aggregatesandattenuatedepositionofamyloidplaquesinaging metabolizingenzymes(MMP2,MMP9,IDE,orneprilysin)in APP/PS1mice.Toinvestigatethispremise,wesubjectedTFEB- TFEB-transduced hippocampi (Fig. 3C). Analogously, the transduced(andcontrol)hippocampaltissuetosequentialho- proteinabundancefortheseenzymeswasunchanged(MMP2, mogenization in PBS followed by guanidine to quantify A(cid:2)in MMP9,andIDE)ormodestlyreduced(neprilysin;Fig.3D,E), PBS-soluble and PBS-insoluble fractions. In addition, we per- suggestingthatregulationofextracellularproteasesisunlikely formedA(cid:2)immunohistochemistryandX-34stainingtoassess to contribute to a reduction in ISF A(cid:2)levels with neuronal A(cid:2)andamyloidload,respectively.Asshown(Fig.4A–C),TFEB TFEBtransduction. transductionresultedina40and50%reductioninsolubleA(cid:2)40 12142•J.Neurosci.,September2,2015•35(35):12137–12151 Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation Figure3. NeuronalTFEBtransductionreducesISFA(cid:2)levels,invivo.A,AssessmentofA(cid:2)levelsbyinvivomicrodialysisin3-month-oldAPP/PS1micetransducedwithAAV8-CMV-FLAG-TFEB (TFEB)andAAV8-CMV-GFP(GFP)withserialhourlymeasurements.N(cid:5)8miceinTFEBgroup,n(cid:5)9miceinGFPgroup.Att(cid:5)0,micewerecontinuallyadministeredCompoundEdirectlytothe hippocampus(200nM,reversemicrodialysis)followedbyhourlysamplingforA(cid:2).Insetshowsmeanabsoluteinvivo“exchangeable”A(cid:2)(eA(cid:2)x-40)concentrationaveragedovera9hperiodbefore drugadministration;*p(cid:8)0.05.B,SemilogplotofdeclineinpercentagebasalISFA(cid:2)levelsduringadministrationofCompoundEinanimalsinA.InsetshowsA(cid:2)half-lifeinthetwogroups;*p(cid:8) 0.05.C–E,Transcriptlevels(C)andproteinabundance[immunoblot(D)withquantitation(E)]forenzymesimplicatedinextracellularA(cid:2)metabolisminhippocampaltissuefromAAV8-CMV-FLAG- TFEB(TFEB)andAAV8-CMV-GFP(GFP)-transducedAPP/PS1mice.N(cid:5)5pergroup;**p(cid:8)0.01.F,G,A(cid:2)40andA(cid:2)42levelsindissectedhippocampaltissuesfromAAV8-CMV-FLAG-TFEB(TFEB)and AAV8-CMV-GFP(GFP)-transducedAPP/PS1miceat3monthsofage.TissuewashomogenizedfirstinPBS(D)theninRIPA(E)quantifiedwithELISAassay.HJ2andHJ7.4antibodieswereusedfor captureA(cid:2)40andA(cid:2)42,respectively,andHJ5.1antibodywasusedfordetection.N(cid:5)6micepergroup;**p(cid:8)0.01. and A(cid:2)42, and 36 and 35% reduction in insoluble A(cid:2)40 and immunostainedamyloidplaqueload(Fig.4G,H).Importantly, A(cid:2)42,respectively,comparedwithcontrols,byanELISAassayto aswehaveobservedpreviouslywithastrocyticexpressionofex- differentiallycapturethetwospeciesbyspecificC-terminalanti- ogenous TFEB (Xiao et al., 2014), the effect of neuronal TFEB bodies.Toconfirmthesefindings,wealsousedanalternateA(cid:2) transduction on amyloid deposition was localized to the hip- peptide capture and detection strategy using a combination of pocampus(seeareaoutlinedwithdottedlineinFig.4DandG antibodies targeting distinct epitopes at the central domain andquantitationinEandH),whichwastargetedforTFEBex- (HJ5.1)andNterminus(HJ3.5),aspreviouslydescribed(Xiaoet pressionbystereotacticinjectionandnotinthecorticalareasthat al.,2014),andobserveda29and31%reductioninthesoluble aredistantfromthesiteofAAVtransduction.Itisinterestingto and insoluble levels of all A(cid:2)species in TFEB-transduced hip- notethatthedegreeofreductioninplaqueloadmirroredthatof pocampi(Fig.4A–C).ThissuggestsauniformreductioninA(cid:2) thereductioninA(cid:2)levels(Fig.4B,C)intheagedmiceandthatof levelsratherthanareductioninspecifictruncatedfragmentswith thereductioninISFsteady-stateA(cid:2)(Fig.3A,B)andhippocam- exogenousTFEBexpression. palA(cid:2)inthepredepositingmice(Fig.3F,G),attestingtoacon- This was accompanied by a 35% reduction in X-34-stained sistenteffectofTFEBonA(cid:2)generationthroughoutthevarious amyloid deposits (Fig. 4D,E) and a 40% reduction in A(cid:2)- stagesofamyloidpathogenesis.ItisalsonoteworthythatTFEB- Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation J.Neurosci.,September2,2015•35(35):12137–12151•12143 transduction had a comparable effect on amyloid plaque load reduction in either sex (Fig. 4F,I), as we have previously ob- served (Xiao et al., 2014).These data suggest that TFEB expressioninneuronsattenuatesamyloidplaquedepositionvia reducingA(cid:2)generation. NeuronalTFEBtransductionreducesAPPandA(cid:2)levelsin wild-typemice ToexcludethepossibilitythatTFEBprimarilyaffectstheprocess- ing of the mutant APP695 protein, which is co-overexpressed withafamilialADriskconferring(cid:2)E9mutantpresenilin-1pro- teinintheAPP/PS1mice(Jankowskyetal.,2004),weexamined theeffectofneuronallytransducedTFEBontheabundanceof endogenous wild-type APP protein and its (cid:3)- and (cid:2)-CTFs in hippocampiofwild-typemiceversusthoseinGFP-transduced hippocampiascontrols.Remarkably,TFEBtransductionledtoa comparabledegreeofreductioninabundanceoftotalendoge- nousAPPandlevelsofthe(cid:3)-and(cid:2)-CTFs(Fig.5A,B)mimicking itseffectinAPP/PS1transgenicmice(Fig.2A,B).Thistranslated intoa27and21%reductioninA(cid:2)40andA(cid:2)42species(Fig.5C), respectively,indicatingthatTFEBalsoaltersthemetabolismof wild-typeAPPproteintoreduceA(cid:2)generation. TFEBexpressionattenuatesA(cid:2)generation,invitro ToconfirmthatTFEBexpressionresultsinreducedA(cid:2)genera- tion, we transfected N2a cells stably expressing APP695 with TFEBorvectorcontrol,andexaminedAPPprocessing.TFEB- transfectedcellsdemonstratedincreasedlysosomeabundance,as indicatedbyupregulationoflysosomalproteins(Figure6A,B) drivenbyeightfoldoverexpressionofTFEB,asweobservedpre- viouslyinC17.2cells(Xiaoetal.,2014).TFEB-transfectedcells demonstratedareductioninAPPabundance,aswellasreduced levelsof(cid:3)-and(cid:2)-CTFsandsAPP(cid:3)inthemedium(Fig.6C,D). ThiswasnotassociatedwithachangeinAPPtranscript(TFEB/ GAPDH)mRNA1.04(cid:7)0.04-foldinTFEBtransfectedvs1.0(cid:7) 0.01-foldincontrol,p(cid:5)0.38,N(cid:5)3pergroup),indicatingthat TFEBstimulatesincreaseddegradationofAPP,whichisconsis- tentwiththeobservationsinTFEB-transducedhippocampi(Fig. 2).ConfocalimagingofadualfluorescentAPPconstruct(San- nerudetal.,2011)alsodemonstratedaqualitativeupregulation oflysosomeabundancewithreductioninfull-lengthAPPprotein in TFEB-transfected cells, along with increased localization of APP in lysosomes (Fig. 6E). Colocalization of full-length APP (bearingbothblueandgreenfluorescenttags)withLysoTracker Red-labeledlysosomes(seearrows)wasqualitativelyobservedto beupregulatedinTFEB-expressingcells(Fig.6E,arrows).This wasassociatedwithreducedA(cid:2)abundance,bothintracellularly (Fig. 7A) and in the medium (Fig. 7B). Importantly, TFEB- transfectedcellsdemonstratedareductioninA(cid:2)appearancein themedium(Fig.7C,D),indicatingthatTFEBprimarilyattenu- atesA(cid:2)generation,andreducedintracellularA(cid:2)levelsarenota resultofincreasedA(cid:2)secretion. Figure4. NeuronalTFEBtransductionreducesamyloidplaqueloadinAPP/PS1mice.A, SchematicrepresentationofspecificantibodiesusedinELISA.B,C,A(cid:2)40andA(cid:2)42levelsin TFEBacceleratesAPPfluxdowntheendosome- dissectedhippocampaltissuesfromAAV8-CMV-FLAG-TFEB(TFEB)andAAV8-CMV-GFP(GFP)- lysosomepathway transducedmice(10monthsofage).TissuewashomogenizedfirstinPBS(solublelevels,B), The intracellular itinerary of APP follows a pathway from the thenin5mMguanidine(insolublelevels,C)quantifiedwithELISAassay.HJ2andHJ7.4anti- Golgi (trans-Golgi network), wherein it is post-translationally bodieswereusedforcaptureA(cid:2)40andA(cid:2)42,respectively,andHJ5.1antibodywasusedfor detection.TotalA(cid:2)levelswerealsomeasuredwithacombinationofHJ5.1antibodyforcapture 4 andHJ3.5antibodyfordetection,asindicatedintheschematic.N(cid:5)8micepergroup;*p(cid:8) 0.05,**p(cid:8)0.01.D,RepresentativeX-34-stainedimagesfromAPP/PS1micetreatedasinA. G,RepresentativeA(cid:2)-immunostainedimagesfrommicetreatedasinB.H,I,Quantificationof Theareaofthehippocampusisoutlinedwithadottedline.E,F,QuantificationofX-34-stained A(cid:2)-stainedplaqueburdeninthehippocampusinmicetreatedasinA(H)andplaqueburden plaqueburdeninthehippocampus(HPC)inmicetreatedasinA(E)andplaqueburdenstratified stratifiedbysex(I).N(cid:5)14(6maleand8female)micepergroup;*p(cid:8)0.05,**p(cid:8)0.01.HPC, by sex (F). N (cid:5) 14 (6 male and 8 female) mice per group;*p (cid:8) 0.05, **p (cid:8) 0.01. Hippocampus;CTX,cortex. 12144•J.Neurosci.,September2,2015•35(35):12137–12151 Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation Figure5. NeuronalTFEBtransductionreduceslevelsofAPP,itsCTFs,andA(cid:2)speciesinwild-typemice.A,B,Immunoblot(A)withquantitation(B)ofAPP,andits(cid:3)-and(cid:2)-CTFsinwild-type mousehippocampitransducedwithAAV8-CMV-FLAG-TFEB(TFEB)orAAV8-CMV-GFP(GFP)at8monthsofage.N(cid:5)3pergroup;*p(cid:8)0.05.C,A(cid:2)40andA(cid:2)42levelsindissectedhippocampal tissueshomogenizedinRIPAbuffer,frommicetransducedasinA.HJ2andHJ7.4antibodieswereusedforcaptureA(cid:2)40andA(cid:2)42,respectively,andHJ5.1antibodywasusedfordetection.N(cid:5) 6pergroup;*p(cid:8)0.05. Figure6. TFEBexpressionincreaseslysosomesandreduceslevelsofAPPanditscleavedCTFs,invitro.A,B,Immunoblots(A)withquantification(B)oflysosomalproteinsin N2a-APP695cellstransfectedwithTFEBorvectorcontrol(for48h).C,D,Immunoblot(C)withquantitation(D)ofAPPandits(cid:3)-and(cid:2)-CTFsandofsAPP(cid:3)fragmentintheoverlying medium,collectedoveradurationof6hincellstreatedasinA.N(cid:5)3pergroup;*p(cid:8)0.05,**p(cid:8)0.01.E,N2acellsweretransfectedwithadualfluorescentAPPconstruct(Sannerud etal.,2011)withorwithouttheTFEBconstructandstainedwithLysoTrackerRed.Asshownintheseconfocalimages,colocalizationoffull-lengthAPP(bothblueandgreentags)with LysoTrackerRed-labeledlysosomes(seearrows)wasqualitativelyincreasedinTFEB-expressingcells(seearrows)comparedwithemptyvector-expressingcells. Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation J.Neurosci.,September2,2015•35(35):12137–12151•12145 al.,2012).Atvarioustimepointspostre- warming, the cells were treated with 2-mercaptoethanesulfonicacidtoremove cell-surface biotinylated APP molecules, permitting assessment of kinetics of ap- pearance and subsequent degradation of total internalized biotin-tagged APP via streptavidin capture and immunoblot- ting.AsshowninFigure8,AandB,cells transfected with TFEB demonstrated reducedlevelsofbothsurfaceandinter- nalizedAPP,suchthattheratioofinter- nalized to surface APP was not altered comparedwithcontrol. To determine whether the reduced steady-statelevelsofAPPareduetoaccel- eratedendocytosisorenhancedfluxinto thelysosomesfordegradation,wedeter- mined the kinetics of intracellular APP appearance in this assay (Fig. 8C,D). In- terestingly,APPwasrapidlyendocytosed and the rate of increase of intracellular APPlevels(fromT(cid:5)1toT(cid:5)7.5min) was not different between TFEB-expre- Figure7. TFEBexpressionattenuatesA(cid:2)generation,invitro.A,B,AbundanceofA(cid:2)40andA(cid:2)42speciesinthecelllysates(A) ssingcellsandcontrols(Fig.8C,D).These andmedium(B)inN2a-APP695cellstransfectedwithTFEBorvectorcontrol.C,D,AccumulationofA(cid:2)40(C)andA(cid:2)42(D)species dataareconsistentwithalackofeffectof intheoverlyingmediumofcellstreatedasinA,overtheindicatedduration.N(cid:5)5pergroup;*p(cid:8)0.05,**p(cid:8)0.01. TFEB on clathrin-mediated endocytosis, consistentwithourpreviousobservations modified after synthesis in the ER, to the plasma membrane (Xiaoetal.,2014).TodeterminewhetherTFEBexpressionaffects whereitcanbecleavedviaanonamyloidogenic(cid:3)-cleavage.Al- the kinetics of other transmembrane proteins taken up by ternatively,APPcanbeendocytosed,andmultiplestudieshave clathrin-mediatedendocytosis,weexaminedtheuptakeofexog- demonstratedthatendocytosisofAPPisessentialforitscolocal- enouslyappliedbiotin-labeledtransferrin.Transferrinbindstothe ization with (cid:2)- and (cid:4)-secretases within endosomes and mul- transferrinreceptorandisendocytosedfollowedbyrecycling tivesicular bodies (formed by trafficking of endosomes into to the cell surface (Ciechanover et al., 1983). The kinetics of intraluminalvesiclesviasequentialactivityofendosomal-sorting intracellular appearance of transferrin was not affected by complexesrequiredfortransport,whicharetheprimarysitesfor TFEB expression (Fig. 8E), confirming that TFEB does not A(cid:2)generationviaamyloidogeniccleavage;RajendranandAnn- accelerateclathrin-mediatedendocytosis. aert, 2012). Studies have suggested that impairment in endo- Incontrast,internalizedAPPlevelsat30minwere32%lower somalflux,likelysecondarytolysosomedysfunction,resultsin in TFEB-transfected cells versus 14% lower in control vector- increasedtransittimewithinthisorganelle,whichincreasesthe transfectedcellsfromtheirrespectivepeaklevelsat7.5min(p(cid:5) propensity for (cid:2)- and (cid:4)-cleavage and, hence, A(cid:2)generation 0.017,n(cid:5)3experimentaltrailspergroup),suggestingthatTFEB (Cataldoetal.,2000;RajendranandAnnaert,2012;Moreletal., acceleratesdegradationofAPP,whichistargetedforlysosomal 2013;PericandAnnaert,2015).Indeed,afterendocytosis,studies degradation,incontrasttotransferrinthatgetsrecycledtothecell have demonstrated that a significant proportion of APP is tar- surface (Ciechanover et al., 1983). Together with the observed getedtothelysosomes(Benowitzetal.,1989;Haassetal.,1992; reductioninAPPanditsvariouscleavedfragments(Fig.6C,D) Caporasoetal.,1994)andAPPlevelsrapidlybuildupinthecells and attenuated A(cid:2)generation (Fig. 7A–D), these observations inthepresenceoflysosomalacidificationinhibitors(Goldeetal., suggest that TFEB expression does not affect APP endocytosis, 1992),suggestingthatlysosomaldegradationdrivesAPPprote- but stimulates increased flux of endocytosed APP down the olysis to preclude formation of A(cid:2)peptides (Caporaso et al., endosome-lysosomepathwaywithoutbuildupofAPPproteinin 1992).GivenpreviousreportsthatTFEBexpressioncanstimu- intracellularcompartments,resultinginitslysosomaldegrada- late endocytosis ( Pen˜a-Llopis et al., 2011), a potential adverse tionandreducedabundanceofAPPatsteadystate. consequenceofTFEBactivationcouldbeincreasedAPPendocy- tosisresultinginenhanced(cid:2)-and(cid:4)-cleavageandacceleratedA(cid:2) TFEBenhanceslysosomaldegradationofAPP generation,asobservedwithotherexperimentalmanipulations ToassesstheroleoflysosomesinAPPdegradation,wetreated tostimulateendocytosis(Grbovicetal.,2003;Schneideretal., TFEB-transfected(andcontrol)cellswithbafilomycinA1toin- 2008;Xiaoetal.,2012).Alternatively,TFEBmaystimulateflux hibit lysosome acidification and evaluated APP abundance. As throughtheendosome-lysosomepathwaytopreventAPPaccu- showninFigure9A,bafilomycintreatmentresultedinincreased mulation in endosomes and stimulate its proteolysis in lyso- APP levels (by 42% compared with control) and largely pre- somes.Toevaluatethesepossibilities,weexaminedtheeffectof vented the TFEB-induced reduction in APP abundance (Fig. TFEBonAPPinternalization.N2a-APP95cellsweretransfected 9A,B).Thiswasassociatedwithincreaseinboth(cid:3)-and(cid:2)-CTF withTFEBorvectorcontrol,andcell-surfaceAPPwaslabeledby levels(Fig.9A,C,D)aswellasincreasedgenerationofA(cid:2)40(Fig. biotinylation at 4°C (to prevent endocytosis), followed by re- 9E)andA(cid:2)42(Fig.9F),asinhibitinglysosomaldegradationof warmingtostimulateitsinternalizationbyendocytosis(Xiaoet APPmakesitincreasinglyavailableasasubstrateforprocessing 12146•J.Neurosci.,September2,2015•35(35):12137–12151 Xiaoetal.•NeuronalTFEBExpressionAttenuatesA(cid:2)Generation Figure8. TFEBexpressionreducessteady-statecell-surfaceandintracellularAPPlevelswithoutalteringitsendocytosis.A,B,Immunoblot(A)andquantitation(B)ofcell-surfaceand internalizedAPPinN2a-APP695cellstransfectedwithTFEBorvectorcontrol,aftercell-surfaceAPPwaslabeledbybiotinylationat4°C(topreventendocytosis)followedbyrewarming(to37°C)to stimulateitsinternalizationbyendocytosis(for10min).Cell-surfaceAPPwasassessedincellsmaintainedat4°C.ToassessinternalizedAPP,cellsweretreatedwith2-mercaptoethanesulfonicacid toremovecell-surfacebiotinylatedAPPmolecules,followedbystreptavidincaptureandimmunoblotting(seeMaterialsandMethods).InsetshowsratioofinternalizedtosurfaceAPP;*p(cid:8)0.05. C–E,Immunoblot(C)andquantitation(D)ofintracellularAPPasafractionofcell-surfaceAPPtodemonstratekineticsofintracellularA(cid:2)incellstransfectedasinAandkineticsofuptakeof biotinylatedtransferrin[immunoblot(C)withquantitation(E)]incellstreatedasinA.N(cid:5)3pergroup.Nostatisticallysignificantdifferenceswereobservedbytwo-wayANOVA. viathesecretases.WenextdeterminedAPPhalf-lifebytreating onstratethatTFEBexpressionacceleratesendogenouspathways cellswithcycloheximidetoinhibitproteinsynthesisandfollow for holo-APP degradation via lysosomes, preventing its amy- APPabundance.AsshowninFigure9,GandH,TFEBtransfec- loidogenicprocessingintoA(cid:2). tionreducedAPPhalf-lifeby42%comparedwithcontrolindi- cating enhanced APP degradation. Intriguingly, the levels of Discussion exogenousTFEBalsodeclinedrapidly,consistentwiththeobser- ContemporaryapproachestoreducingA(cid:2)generationinneurons vations that activated TFEB is degraded rapidly (Roczniak- havefocuseduponshiftingAPPprocessingtononamyloidogenic Ferguson et al., 2012) possibly via the ubiquitin-proteasome pathwaysorsortingAPPawayfromtheendosomeswhereamy- pathway(Maiaetal.,2015).Itwasalsointerestingtonotethatthe loidogenic cleavage takes place (Haass et al., 2012). Our study abundanceofAPPinbafilomycin-treatedTFEB-expressingcells demonstratestheefficacyofacceleratingholo-APPdegradation waslowerthantherespectivelevelsinvectorcontrols(Fig.9B), inlysosomesasastrategytoreduceA(cid:2)generation.Indeed,exog- againpointingtothereductioninsteady-statelevelsofAPPwith enousTFEBexpressionstimulateslysosomebiogenesisinvitro, TFEB expression, before application of bafilomycin to inhibit reducesfull-lengthAPPanditscleavedfragments,attenuatesA(cid:2) lysosomeacidification.Expectedly,thisresultedinarelativede- generationandreleaseinthemedium(Figs.6,7),andmarkedly clineintheCTFsandA(cid:2)peptidesinthissetting(Fig.9C–F). shortensAPPhalf-life(Fig.9)inalysosome-dependentmanner. Asdescribedpreviously(Caporasoetal.,1992),inhibitionof These effects are likely secondary to acceleration of lysosomal lysosomeacidificationwithbafilomycinA1prolongedAPPhalf- degradation of endocytosed APP, as suggested by reduction of life,demonstratingthatasignificantfractionofAPPisdegraded steady-state APP at both the surface and within intracellular viathelysosomes(Fig.9I–K).Importantly,APPhalf-lifewasalso compartments (Fig. 8), and reduced APP half-life with TFEB markedlyprolongedby61%inthepresenceofbafilomycinA1in expression (Fig. 9). Mirroring these in vitro findings, AAV- TFEB-transfectedcells(Fig.9L).Insummary,thesestudiesdem- mediated neuronal TFEB transduction in the hippocampus of

Description:
X Danielle L. Tripoli,1 X Leah Czerniewski,1 Andrea Ballabio,5 John R. Cirrito,1 X Abhinav Diwan,2,3,4* and. X Jin-Moo Lee1*. 1Department of
See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.