• Nem Talált Eredményt

species and expression of matrix metalloproteinases generation by suppressing apoptosis of reactiveoxygen/nitrogen Activation protects of PPAR cardiac ␤ / ␦ myocytes from oxidativestress-induced Pharmacological Research

N/A
N/A
Protected

Academic year: 2022

Ossza meg "species and expression of matrix metalloproteinases generation by suppressing apoptosis of reactiveoxygen/nitrogen Activation protects of PPAR cardiac ␤ / ␦ myocytes from oxidativestress-induced Pharmacological Research"

Copied!
9
0
0

Teljes szövegt

(1)

ContentslistsavailableatScienceDirect

Pharmacological Research

jo u r n al hom e p ag e :w w w . e l s e v i e r . c o m / lo c a t e / y p h r s

Activation of PPAR ␤ / ␦ protects cardiac myocytes from oxidative stress-induced apoptosis by suppressing generation of reactive

oxygen/nitrogen species and expression of matrix metalloproteinases

Eleftheria Barlaka

a

, Anikó Görbe

b,d

, Renáta Gáspár

b

, János Pálóczi

b

, Péter Ferdinandy

c,d

, Antigone Lazou

a,∗

aLaboratoryofAnimalPhysiology,SchoolofBiology,AristotleUniversityofThessaloniki,Greece

bCardiovascularResearchGroup,DepartmentofBiochemistry,UniversityofSzeged,Hungary

cDepartmentofPharmacologyandPharmacotherapy,SemmelweisUniversity,Budapest,Hungary

dPharmahungaryGroup,Szeged,Hungary

a r t i c l e i n f o

Articlehistory:

Received2November2014

Receivedinrevisedform15March2015 Accepted15March2015

Availableonline28March2015

Keywords:

Cardiacmyocytes Oxidativestress Apoptosis

Matrixmetalloproteinases PPAR␤/␦

Caspase3 Bcl-2

Chemicalcompoundsstudiedinthisarticle:

Hydrogenperoxide(PubChemCID:784) GW0742(PubChemCID:9934458) GSK0660(PubChemCID:46233311)

a b s t r a c t

Heartfailurestillremainsoneoftheleadingcausesofmorbidityandmortalityworldwide.Amajor contributingfactorisreactiveoxygen/nitrogenspecies(RONS)overproductionwhichisassociatedwith cardiacremodelingpartlythroughcardiomyocyteapoptosis.Peroxisomeproliferator-activatedreceptors (PPARs)areligand-activatedtranscriptionfactorsthatbelongtothenuclearreceptorsuperfamilyand havebeenimplicatedincardioprotection.However,themolecularmechanismsarelargelyunexplored.In thisstudywesoughttoinvestigatethepotentialbeneficialeffectsevokedbyactivationofPPAR␤/␦under thesettingofoxidativestressinducedbyH2O2inadultratcardiacmyocytes.TheselectivePPAR␤/␦ago- nistGW0742inhibitedtheH2O2-inducedapoptosisandincreasedcellviability.Inaddition,generation ofRONSwasattenuatedincardiacmyocytesinthepresenceofPPAR␤/␦agonist.Theseeffectswereabol- ishedinthepresenceofthePPAR␤/␦antagonistindicatingthattheeffectwasthroughPPAR␤/␦receptor activation.TreatmentwithPPAR␤/␦agonistwasalsoassociatedwithattenuationofcaspase-3andPARP cleavage,upregulationofanti-apoptoticBcl-2andconcomitantdownregulationofpro-apoptoticBax.

Inaddition,activationofPPAR␤/␦inhibitedtheoxidative-stress-inducedMMP-2andMMP-9mRNA upregulation.ItisconcludedthatPPAR␤/␦activationexertsacytoprotectiveeffectinadultratcardiac myocytessubjectedtooxidativestressviainhibitionofoxidativestress,MMPexpression,andapopto- sis.OurdatasuggestthatthenovelconnectionbetweenPPARsignalingandMMPdown-regulationin cardiacmyocytesmightrepresentanewtargetforthemanagementofoxidativestress-inducedcardiac dysfunction.

©2015ElsevierLtd.Allrightsreserved.

1. Introduction

Cardiovasculardiseasesstillremainoneoftheleadingcauses ofmortalityandmorbidityworldwide.Overthepastyears,clinical andexperimentalstudieshaveprovidedsubstantialevidencethat oxidativestress,definedasanimbalancebetweenfreeradicalpro- ductionandtheirdetoxificationbyantioxidants,isanimportant contributingfactorinthepathogenesisofavarietyofcardiovas- culardiseasesincludingischemicheartdiseaseandheartfailure

Correspondingauthorat:LaboratoryofAnimalPhysiology,SchoolofBiology, AristotleUniversityofThessaloniki,Thessaloniki54124,Greece.

Tel.:+302310998381;fax:+302310998269.

E-mailaddress:lazou@bio.auth.gr(A.Lazou).

[1,2]. Excessivegeneration of reactive oxygen/nitrogen species (RONS) causescellular dysfunction,protein and lipidperoxida- tion,DNAdamage,activationofmatrixmetalloproteinases(MMPs) andcanleadtoirreversiblecelldamageandcelldeath.Thecon- trolofmyocytelossthroughsuppressionofcelldeathpathways representsapromisingstrategytopreventcardiacdiseases.Ter- minally differentiated adult cardiac myocytes die primarily by apoptosis or necrosis[3]. Mitochondrial damage, cytochrome c releaseandactivationofcysteineproteasescaspase9and3have beenimplicatedinapoptoticpathways.TheBcl-2familyproteins, which containboth pro-apoptotic and anti-apoptoticmembers, havebeenrecognizedasimportantmodulatorsofcardiacmyocyte apoptosisasin othercells [4,5].Bcl-2familyproteinsactatthe mitochondriaeitherasheterodimersorashomodimerstoregu- latecytochromecwhereasthedynamicequilibriumbetweensuch http://dx.doi.org/10.1016/j.phrs.2015.03.008

1043-6618/©2015ElsevierLtd.Allrightsreserved.

(2)

E.Barlakaetal./PharmacologicalResearch95–96(2015)102–110 103

complexesappearstodeterminethepredispositiontoapoptosis [6].

Thezinc-dependentendopeptidases,matrixmetalloproteinases (MMPs),which havebeenknownfor theirability toproteolyse extracellular matrix proteins, play an important role in tissue remodelingassociatedwithvariousphysiologicalandpathological processesincludingchronicheartfailure[7].Itisnowrecognized thatMMPscanalsohavenon-matrixrelatedintracellularprotein targetsandcanregulatediversecellularfunctionscontributingto cardiacdysfunction,suchasoxidative stress-inducedcell death [8–10].Thus,MMPsareconsideredtobeviabledrugtargetsinthe therapyofcardiacpathologies.

Peroxisomeproliferatoractivatedreceptors(PPARs)areligand activatedtranscription factorsthat belongtothesuperfamilyof nuclearhormonereceptorsandinclude3members(␣,␤/␦,␥)with tissuespecificbutoverlappingexpressionpatterns.Uponactivation bytheirnaturalorpharmaceuticalspecificligands,PPARsregulate theexpressionofgenesmostlyinvolvedinlipidandcarbohydrate metabolism,energyproductionandinflammation[11].Ithasbeen alsoshownthatPPARsmayplayaroleinregulatingtranscriptional expressionofantioxidants,suchasCu/Zn-superoxidedismutase (SOD1),manganese superoxide dismutase (SOD2),and catalase [12,13].Furthermore,emergingevidenceindicatesthatactivation ofPPARs,inparticularPPAR␣orPPAR␥,mayexertcardioprotec- tiveeffects beyondtheirmetaboliceffects,decreasinginfarction afterI/Randpreventingcardiacremodelingandfailure[14–17].

However,themechanismsunderlyingtheseprotectiveeffectsare largelyunexplored,anditisstillamatterofdebatewhethercar- dioprotectionisattributabletothemodulationofcardiacenergy metabolismortoantioxidativeandanti-inflammatoryeffectsof PPARs[18,19].AlthoughPPAR␤/␦isthepredominantsubtypein theheart,itistheleaststudiedmemberofthePPARfamily.Car- diacPPAR␤/␦deletioninmiceresultedindepressedbioenergetics, cardiachypertrophy,andcongestiveheartfailure[20]whileithas beenshownthatPPAR␤/␦isessentialfortheadulthearttomaintain mitochondrialcapacityandoxidativemetabolism[21].

GiventheemergingroleofPPAR␤/␦inregulatingmitochondrial biogenesisandmetabolism,wehypothesizedthatPPAR␤/␦acti- vationmayameliorateoxidativestress-inducedcardiacmyocyte damage.WeusedthehighlyselectiveandpotentPPAR␤/␦agonist GW0742[22]andaspecificPPAR␤/␦antagonist,GSK0660[23]as pharmacologicaltoolstoevaluatetheeffectsofreceptoractivation inH2O2-inducedcardiacmyocyteapoptosisandexplorethecon- tributingmechanisms.WedemonstratethatPPAR␤/␦activationin cardiacmyocytesleadstotheinhibitionofRONSgeneration,down- regulationofMMP-2andMMP-9gene expressionandprevents apoptosis.

2. Materialsandmethods 2.1. Animals

MaleWistarratsweighingbetween250and300gwereused.

Animalsreceivedpropercareincompliancewiththe“Guidelines for the Care and Use of Laboratory Animals” publishedby US NationalInstitutesofHealth(NIHpublicationNo85-23,revised 1996) and the “Principlesof laboratoryanimal care” published by theGreek Government (160/1991)based on EU regulations (86/609).Allsurgerywasperformedundersodiumpentobarbital anesthesia,andalleffortsweremadetominimizesuffering.

2.2. Isolationofadultratcardiacmyocytesandtreatments

Ventricularmyocyteswereisolatedbycardiacretrogradeaor- ticperfusionandcollagenase treatmentas describedpreviously

[24]. Cells were finally resuspended in incubation medium, in which added Ca2+ was gradually increased to 1mM. Prepara- tionswereconsideredsatisfactoryonlyiftheyieldofrod-shaped cells was more than 70%. Experiments were performed30min afterdissociation.Cardiacmyocyteswereeitherleftuntreated,or theywerepre-treatedwiththePPAR␤/␦agonistGW0742(50nM) (Sigma–AldrichChemieGmbH,Taufkirchen,Germany)for10min beforeexposuretoH2O2 (0.1mM).Whenthe PPAR␤/␦antago- nistGSK0660(0.5␮M)(TocrisBioscience,Bristol,UK)wasused, itwasadded10minbeforetheadditionoftheagonistGW0742.

TheconcentrationofH2O2toinduceapoptosiswaschosenaccord- ingtoourpreviousstudies[24–26].Theoptimalconcentrationof PPAR␤/␦agonistandantagonistwerechosenaccordingtoprelim- inaryexperiments.Preliminaryexperimentsalsoestablishedthat theconcentrationsoftheagonistandantagonistuseddidnotaffect theviabilityofcardiacmyocytes.Theexperimentalprotocolsare summarizedinFig.1.

2.3. Determinationofcellviability

Cellviabilitywasdeterminedusingthe3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay which assesses mitochondrialactivityin livingcells. Cardiacmyocytes (105/sample)wereeitherleftuntreatedortheywerepretreated with PPAR␤/␦ agonist and/or antagonist (10min), followed by exposuretoH2O2for3h.Tetrazoliumbromideataconcentration 0.1mg/mlwasadded10minbeforetheendofincubation.Atthe endofincubation,1mlofethanolwasaddedfor10mintodissolve formazanprecipitate.Absorbancewasmeasuredat570nmusinga microplateautoreader.Cellviabilitywascalculatedaspercentage of control OD. Negativecontrols wereused to ensure that the drugsusedforthecelltreatmentsdidnotinterfereontheirown withtheformationofformazan.

2.4. Measurementoflactatedehydrogenase(LDH)

LDH release into the cell culture medium was used as an indicatorofcelldeath. LDHactivitywasdeterminedbyspectro- photometric analysis measuringthe decrease in absorbance at 340nmresultingfromtheoxidationofNADH.Attheendoftreat- ments,cellculturemediumwascollectedand LDHactivitywas measuredafteradditionofNADH(0.17mM)andsodiumpyruvate (1mM).LDHactivitywasexpressedasmunits/mgafterdetermi- nationofproteincontentpersamplebyBradfordassay.

2.5. Evaluationofapoptosis

Apoptosiswasdeterminedbystainingcells withAnnexinV- FITCandpropidiumiodideforflowcytometricanalysis(Biolegend Europe).Briefly,aftertreatments,cells werewashedtwicewith coldphosphate-bufferedsalineandresuspended.Thecells were incubatedwithAnnexinV-FITCandpropidiumiodidefor15min inthedark.Thesampleswerethen subjectedtoflowcytomet- ricanalysistodetectthepercentage ofapoptotic (FITC-stained) and necrotic (PI-stained) cells in a given population. Fluores- cence was measured on a CyFlow ML flow cytometer (Partec GmbH,Germany).Preliminary experimentsestablishedthat the agonistGW0742 ortheantagonist GSK0660usedfor thetreat- mentofcellsdidnotinterfereontheirowninthemeasurementof fluorescence.

2.6. RONSdetection

Intracellular RONS production was evaluated using the 5- (and-6)-chloromethyl-2,7-dichlorodihydrofluorescein diacetate (CM-H2DCFDA) (Sigma–Aldrich Chemie GmbH, Taufkirchen,

(3)

Fig.1.Diagramofexperimentalprotocolfordrugadministrationandinductionofoxidativestressincardiacmyocytes.Cardiacmyocyteswereeitherleftuntreated(control) ortheywerestimulatedwithH2O2(0.1mM).Whenused,PPAR␤/␦agonist(GW,50nM)wasaddedfor10minbeforestimulationwithH2O2.WhenthePPAR␤/␦antagonist wasused(GSK,0.5␮M),itwasadded10minbeforetheadditionofPPAR␤/␦agonistGW0742.

Germany). In brief, cardiac myocytes were loaded with CM- H2DCFDA (5␮g/ml) and then, they were either left untreated ortheywerepretreatedwiththePPAR␤/␦agonistGW0742 for 10minand/ortheantagonistGSK0660(0.5␮M),inthepresence orintheabsenceofH2O2(0.1mM)for3h.RONS-inducedfluores- cencewasdetectedbyflowcytometry(CyFlowML,PartecGmbH, Germany).PreliminaryexperimentsestablishedthatGW0742or GSK0660didnotinterfereontheirownin themeasurement of fluorescence.

2.7. IsolationofRNAandquantitativePCR(qPCR)

Total RNA was extracted using TRI Reagent (Molecular Research Center, Cincinnati, OH, USA) according to the manu- facturer’sinstructions. TheRNA was resuspended in 0.1% (v/v) diethylpyrocarbamate-treated water and its concentration was determined by absorbance at 260nm. The integrity of isolated RNAwasverifiedona 1%agarosegel.For cDNAsynthesis, l␮g oftotal RNA wasdenaturedin thepresence ofrandom 9-mers (Takara,Japan) ina reactionvolumeof10␮lat65C for5min.

RNAwasreverse-transcribedwithPrimeScriptReverseTranscrip- tase enzyme (Takara, Japan), dNTP’s (HyTest Ltd., Finland) and HumanPlacentalRibonuclease inhibitor(HTBiotechnologyLtd., Cambridge,UK). Thereaction,intotalvolumeof20␮l,wascar- riedoutfor1hat37C,andwasterminatedbyheatinactivation ofthereversetranscriptaseat95Cfor5min.qPCRanalysiswas performedusing a Real-TimePCR System(Applied Biosystems) withaspecificsetofprimersforeachgene(VBCGenomics).The forwardandreverseprimersusedforgeneexpressionwerethe following:

Catalase: For:CCTGTGAACTGTCCCTACCG

Rev:ACCCAGTCCCATGCTCTCTC

MMP-2: For:TTGATGACGATGAGCTGTGG

Rev:CTGCTGTATTCCCGACCATT

MMP-9: For:CCGTGGTCCCCACTTACTTT

Rev:CCGTGGTGCAGGACAAATAG

␤-actin: For:GCCCTGAGGCACTCTTCCA Rev:CGGATGTCCACGTCACACTTC

Eachreaction mixcontained 5␮l KAPASYBR FAST qPCR Kit (KapaBiosystems),0.3␮loligonucleotides(10pmoleach offor- wardand reverse primers)and 2␮lcDNA (diluted1:10). qPCR analysisof␤-actinwasperformedasanendogenouscontrol.PCR conditionswere95Cfor20min,followedby40cyclesof95C for 3sand60C for 30min.FollowingqPCR, dissociationcurve analysiswasroutinelyperformedtocheckforaberrantamplifica- tionproducts(e.g.primers-dimers).Relativechangesinexpression werecalculatedusingthe2(ct)method.

2.8. Preparationofwholecellextracts

Cardiac myocytes were lysed at 4C in a buffer containing 20mM ␤-glycerophosphate, 20mM HEPES pH 7.5, 50mM NaF, 2mM EDTA, 0.2mM Na3VO4, 10mM benzamidine, 5mM DTT, 0.3mMPMSF, 0.2mMleupeptin,0.01mM E64and1%(v/v)Tri- ton X-100. Lysateswere incubated onice for 10min and then centrifugedat4C,for15minat10,000×g.Supernatantswerecol- lected,andtotalproteinwasmeasuredusingtheBradfordassay (BioRad,Hercules,CA,USA).

2.9. Immunoblotting

Total protein extracts were boiled with 0.33vol of SDS- polyacrylamidegelelectrophoresissamplebuffer(10%SDS(w/v),

(4)

E.Barlakaetal./PharmacologicalResearch95–96(2015)102–110 105

13%glycerol(v/v),300mMTris–HCl,pH6.8,130mMdithiothre- itoland0.2%bromophenolblue(w/v).Proteinswereseparatedby SDS-PAGEon10%acrylamide,0.275%(w/v)bis-acrylamideslabgels andtransferredelectrophoreticallyontonitrocellulosemembranes (0.45␮m).Nonspecificbindingsiteswereblocked(30minatroom temperature)with5% (w/v)nonfatmilk powderinTBST buffer [20mMTris–HClpH7.5,137mMNaCl,0.1%(v/v)Tween20].Mem- braneswereincubated(overnight,4C)withprimaryantibodies dilutedinTBSTbuffercontaining5%(w/v)bovineserumalbumin, thenwashedinTBSTbuffer(3×5min,roomtemperature).Primary antibodiesagainstBcl-2(#2876),cleavedcaspase-3(#8698),PARP (#9542),␤-actin(#8457),␣-tubulin(#2125)(CellSignaling,Bev- erly,MA,USA)andBax(sc-7480,SantaCruzBiotechnology,Texas, USA)wereusedatvariousdilutions.Membraneswereincubated (60min,roomtemperature) withhorseradishperoxidase(HRP) conjugatedsecondaryantibodies(1/5000)inTBSTbuffercontain- ing1%(w/v)non-fatmilkpowder,andtheywerethenwashedin TBSTbuffer(3×5min,roomtemperature).Proteinsweredetected byenhancedchemiluminescence(CellSignaling,Beverly,MA,USA) andquantifiedbyscanningdensitometry.

2.10. Statisticalanalysis

Data are presented as mean±SEM of n independent exper- iments using different cell isolations from different animals.

Statisticalanalyses(ANOVAwithDunnett’scomparisontest)were performedwithsignificancetakenasbeingestablishedatp<0.05.

3. Results

3.1. ActivationofPPARˇ/ıwiththeagonistGW0742increases cellviabilityandattenuatesLDHreleaseduringexposureof cardiacmyocytestoH2O2

WehavepreviouslyshownthatH2O2,atconcentrationshigher than0.05mM,decreasesviabilityandinducesapoptosisincardiac myocytes[24–26].Consistentwiththesestudies,exposureofcar- diacmyocytesto0.1mMH2O2for3hdecreasedtheviabilityof cardiacmyocytesbyalmost40% andcaused analmostfourfold increaseinLDHactivitymeasuredintheculturemedium(Fig.2A andB).Pre-treatmentofcardiacmyocyteswiththePPAR␤/␦ago- nist, GW0742, significantly enhanced survival while attenuated LDH release. Inorder todeterminewhetherthis is a PPAR␤/␦- mediatedeffect,thespecificantagonistofPPAR␤/␦,GSK0660,was used. As shown in Fig. 2A, the PPAR␤/␦ agonist GW0742 was unabletoincreasecellviabilityin thepresence ofthePPAR␤/␦

antagonistGSK0660indicatingthatthiseffectoncardiacmyocyte survivalisdependentofPPAR␤/␦activation.Similarly,theantag- onistabolishedtheprotectiveeffectofPPAR␤/␦agonistonLDH release(Fig.2B).NeithertheagonistGW0742northeantagonist GSK0660hadanyeffectoncellsurvivalorLDHactivitywhenused alone.

3.2. ActivationofPPARˇ/ıwiththeagonistGW0742protects cardiacmyocytesfromH2O2-inducedapoptosis

Consistent with previous data [24–26] exposure of cardiac myocytesto0.1mMofH2O2 resultedinalmost50%increasein therate ofapoptosis (Fig. 3A)whereas pre-treatment withthe PPAR␤/␦agonist,GW0742,significantlyattenuatedapoptosis.In thepresenceofthePPAR␤/␦antagonistGSK0660,thePPAR␤/␦ago- nistGW0742wasunabletoattenuatecardiacmyocyteapoptosis indicatingthattheprotectiveeffectoftheagonistisdependenton PPAR␤/␦activation.Tocorroboratefurthertheseresults,caspase- 3 activation and poly ADP-ribose polymerase (PARP) cleavage were alsoevaluated under thesame conditions. Theactivation of caspase-3, the primary executioner of cell death byproteo- lytic processing of pro-caspase, serves as an early marker of apoptosisinvariouscelltypes.Theactivationofcaspasesresults in proteolyticcleavage of numerous substratesincluding PARP, a nuclear enzyme involved in DNA repair that is considered to be a hallmark of apoptosis [27]. As shown in Fig. 3B and C, pre-treatmentof cardiacmyocyteswiththePPAR␤/␦ agonist GW0742inhibitedH2O2-inducedcaspase-3andPARPcleavage,an effectthatisreversedinthepresenceofthePPAR␤/␦antagonist GSK0660.

3.3. ActivationofPPARˇ/ıwiththeagonistGW0742protects cardiacmyocytesfromH2O2-inducedoxidativestress

Given the protective effect of PPAR␤/␦ activation on car- diacmyocyteviability,andapoptosis,wesoughttoexaminethe potentialof PPAR␤/␦ agonistGW0742 toameliorateintracellu- lar generation of RONS in cardiac myocytes.Cells were loaded withthefluorophoreCM-H2DCFDAbeforetreatmentwithH2O2, and/orPPAR␤/␦agonistGW0742andfluorescencewasdetermined withflowcytometry.Asexpected,exposureofcardiacmyocytes toH2O2 resulted inincreased generationof RONSindicative of oxidativestress(Fig.4).However,additionofthePPAR␤/␦agonist completelyabolishedtheincreaseofRONSinducedbyH2O2.The inhibitoryeffectofPPAR␤/␦agonistGW0742onintracellularRONS productionwasreversedwhentheantagonistGSK0660wasadded,

Fig.2.Effectofpre-treatmentofcardiacmyocyteswithPPAR␤/␦agonistGW0742onviabilityandH2O2-inducedLDHrelease.Cardiacmyocyteswereeitherleftuntreatedor theywerepretreatedwithPPAR␤/␦agonistGW0742(GW,50nM)and/orPPAR␤/␦antagonistGSK0660(GSK,0.5␮M).Thentheywereincubatedintheabsenceorpresenceof H2O2(0.1mM)for3h.(A)CellviabilitywasmeasuredusingtheMTTmethod.(B)LDHactivitywasevaluatedbyspectrophotometricmeasurement.Resultsaremean±SEM of4–5independentexperiments.*p<0.05comparedwithcontrol,#p<0.05comparedwithH2O2,ˆp<0.05comparedwithcellstreatedwithH2O2andPPAR␤/␦agonist GW0742.

(5)

Fig.3. Effectofpre-treatmentofcardiacmyocyteswithPPAR␤/␦agonistGW0742onH2O2-inducedapoptosislevelsandcleavedcaspase-3andPARP.Cardiacmyocyteswere eitherleftuntreatedortheywerepretreatedwithPPAR␤/␦agonistGW0742(GW,50nM)and/orPPAR␤/␦antagonistGSK0660(GSK,0.5␮M).Thentheywereincubatedin theabsenceorpresenceofH2O2(0.1mM)for3h.(A)Fortheevaluationofapoptosis,cellswereloadedwithannexin/PIandfluorescencewasdeterminedbyflowcytometry.

(B,C)Cellextractswereimmunoblottedforcleavedcaspase-3andPARP.Equalloadingwasverifiedbyimmunoblottingfor␣-tubulin.(B)Representativeblotsareshown.

(C)Blotswerequantifiedusingscanningdensitometryandplotted.Resultsaremean±SEMof3experimentswithindependentcellpreparations.*p<0.05comparedwith control,#p<0.05comparedwithcellstreatedonlywithH2O2,ˆp<0.05comparedwithcellstreatedwithH2O2andPPAR␤/␦agonistGW0742.

verifyingthatthiseffectismediatedthroughPPAR␤/␦activation.

Furthermore,themRNAlevelsofcatalase,whichisknowntobe aPPAR␤/␦targetgene[28],werealsomeasuredunderthesame conditions(Fig.5).Catalasewasup-regulatedincardiacmyocytes thatwerepre-treatedwithPPAR␤/␦agonistGW0742ascompared withthenon-treatedcellswhereasadditionofthePPAR␤/␦antag- onistabolishedthiseffectindicatingtheactivationofthereceptor.

Takentogethertheseexperimentspointtowardtheantioxidant roleofPPAR␤/␦activation.

3.4. PPARˇ/ıactivationmodulatesBcl-2familyprotein expressionduringcardiacmyocyteoxidativestress

WenextdeterminedwhethertheprotectiveeffectofPPAR␤/␦ activationcanbeattributabletoupregulationofBcl-2protein.The Bcl-2family proteinsarekeycomponentsoftheapoptoticpro- cessin cardiacmyocytesand theratioBcl-2/Baxiswidelyused asamarkerofapoptosis[24,25].We,thus,determinedthepro- teinlevelsoftheanti-apoptoticBcl-2andthepro-apoptoticBax.

(6)

E.Barlakaetal./PharmacologicalResearch95–96(2015)102–110 107

Fig.4.Effectofpre-treatmentofcardiacmyocyteswithPPAR␤/␦agonistGW0742onRONSgeneration.Cardiacmyocyteswerepre-loadedwith5-(and-6)-chloromethyl- 2,7-dichlorodihydrofluoresceindiacetate(CM-H2DCFDA)andthentheywereeitherleftuntreatedortheywerepretreatedwithPPAR␤/␦agonistGW0742(GW,50nM) and/orPPAR␤/␦antagonistGSK0660(GSK,0.5␮M).ThentheywereincubatedintheabsenceorpresenceofH2O2(0.1mM)for3h.ChangesinDCFfluorescencewere determinedbyflowcytometry.Resultsaremean±SEMof5independentexperiments.*p<0.05comparedwithcontrol,#p<0.05comparedwithtreatmentwithH2O2,

ˆp<0.05comparedwithcellstreatedwithH2O2andPPAR␤/␦agonistGW0742.

Fig.5. Effectofpre-treatmentofcardiacmyocyteswithPPAR␤/␦agonistGW0742 oncatalasemRNAexpression.Cardiacmyocyteswereeitherleftuntreatedorthey werepretreatedwithPPAR␤/␦GW0742(GW,50nM)and/orPPAR␤/␦antagonist GSK0660(GSK,0.5␮M).Thentheywereincubatedintheabsenceorpresenceof H2O2(0.1mM)for3h.CatalasemRNAlevelsweredeterminedbyreal-timeRT-PCR andwerenormalizedtothemRNAlevelsofthehousekeepinggene␤-actin.Results aremean±SEMof5independentexperiments.*p<0.05comparedwithcontrol,

#p<0.05comparedwithtreatmentwithH2O2,ˆp<0.05comparedwithcellstreated withH2O2andPPAR␤/␦agonistGW0742.

AsshowninFig.6,treatmentofcardiacmyocyteswithPPAR␤/␦ agonistfollowedbyH2O2resultedinincreasedexpressionlevels ofanti-apoptoticBcl-2comparedwithcellsthatwereexposedto H2O2alone.Inasimilarfashion,theH2O2-inducedincreaseinBax wasattenuatedbythePPAR␤/␦agonist.Theeffectoftheagonist GW0742ontheexpressionofBcl-2familyproteinswasreversed inthepresenceoftheantagonistGSK0660,indicatingthatthisis aPPAR␤/␦-mediatedeffect.Takentogethertheseresultssuggest thatactivationofPPAR␤/␦leadstoincreasedresistancetoH2O2- inducedcardiacmyocyteapoptosisthroughupregulationofBcl-2 andconcomitantdownregulationofBax.

3.5. PPARˇ/ıactivationleadstoinhibitionofMMP-2andMMP-9 mRNAexpressionincardiacmyocytesexposedtoH2O2

Severalstudieshavedocumentedtheroleofmatrixmetallo- proteinases(MMPs)incardiacremodelingandthedevelopment

ofcardiacdisease[29].Furthermore,recentevidenceimplicates MMPsintheproteolyticregulationofseveralintracellularproteins inmyocardialoxidativestressinjuryleadingtocardiacdysfunc- tionandcelldeath[30].Thus,wedeterminedtheeffectofPPAR␤/␦ activationonMMP-2andMMP-9mRNAexpression.Asexpected, exposureofcardiacmyocytestoH2O2resultedinincreasedmRNA expression of both MMP-2 and MMP-9 (Fig. 7A and B). Pre- treatment of cardiac myocytes with PPAR␤/␦ agonist GW0742 resultedinattenuationofMMP-2andMMP-9mRNAlevels(Fig.7A andB).ThiseffectwasreversedbytheadditionofPPAR␤/␦antag- onist.

4. Discussion

Inthisstudy,wefocusonthepotentialbeneficialroleofPPAR␤/␦ againstoxidativestressinduceddysfunctionincardiacmyocytes.

WeprovideevidencethatPPAR␤/␦activationbyitsspecificagonist GW0742protectscardiacmyocytesfromapoptosisviasuppression ofRONSgenerationandMMPgeneexpression.

TheroleofPPARsinthepathogenesisofvariouscardiovascular diseaseshasnotbeenfullyelucidatedandremainsamatterofcon- troversy.AlthoughPPAR␤/␦isthelessstudiedisoform,especially intheheart,thereismountingevidencesupportingitscardiopro- tectiverole.PPAR␤/␦isdownregulatedinratheartafterI/R[14]

whereasconditionalcardiac-specificdeletionofPPAR␤/␦results inincreasedcardiaclipidaccumulationandcardiomyopathy[20].

Furthermore, PPAR␤/␦ agonists arecardioprotective in the set- tingofI/Rasmanifestedbyareducedinfarctsizeandimproved postischaemicrecoveryofcontractilefunctionindifferentanimal models[31,32,Lazouetal.unpublisheddata].Inaddition,activa- tionofPPAR␤/␦hasbeenrelatedtoinhibitionofhypertrophyand inflammationthroughthesuppressionofNF-␬Bandinflammatory cytokines[33–35].

H2O2, as a physiologically relevant form of oxidative stress, induces cardiac myocyte apoptosis [24–26]. In line with a cardioprotectiveroleofPPAR␤/␦,wedemonstratethatpretreat- ment of cardiac myocytes with the PPAR␤/␦ selective agonist GW0742increasescellviabilityandinhibitsH2O2-inducedapo- ptosis(Figs.2and3)Furthermore,pretreatmentwiththeselective

(7)

Fig.6.PPAR␤/␦activationmodulatesBcl-2familyproteinexpressionduringoxidativestress.Cardiacmyocyteswereeitherleftuntreatedortheywerepretreatedwith PPAR␤/␦agonistGW0742(GW,50nM)and/orantagonistGSK0660(GSK,0.5␮M).ThentheywereincubatedintheabsenceorpresenceofH2O2(0.1mM)for3h.Cellextracts wereimmunoblottedforBcl-2andBax.Equalloadingwasverifiedbyimmunoblottingfor␤-actin.Representativeblotsareshown(upperpanels).Blotswerequantified usingscanningdensitometryandplotted(lowerpanels).Resultsaremean±SEMof3–4experimentswithindependentcellpreparations.*p<0.05comparedwithcontrol,

#p<0.05comparedwithcellstreatedonlywithH2O2,ˆp<0.05comparedwithcellstreatedwithH2O2andPPAR␤/␦agonistGW0742.

PPAR␤/␦ antagonist GSK0660 abolishes theprotection afforded byGW0742,andthisfindingimpliesthattheobservedbeneficial effectsofGW0742areindeedsecondarytotheselectiveactivation ofthePPAR␤/␦receptor.Thereareseveralpotentialmechanisms bywhichthePPAR␤/␦agonistmayexertitsanti-apoptoticeffect.

Our results showthat additionof thePPAR␤/␦ agonist in car- diacmyocytes amelioratesoxidative stressasevidenced bythe inhibitionofRONSgeneration(Fig.4).Manyimportantendoge- nousanti-oxidantssuchasCu/Zn-superoxidedismutase(SOD1), manganesesuperoxidedismutases (SOD2) and catalasecontain functionalPPARresponseelements(PPREs)intheirpromoteranda

roleofPPAR␤/␦inthetranscriptionalregulationoftheseenzymes hasbeenpreviouslyreported[21,28,36].Upregulationofthecata- lase gene wasobserved in the presence of PPAR␤/␦agonist in cardiacmyocytes(Fig.5).Althoughcatalaseactivitywasnotmea- suredinthisstudy,increasedcatalasegeneexpressionmayindicate anincreasedanti-oxidantcapacity.PPAR␤/␦hasbeensuggestedto inhibitROSgenerationthroughtheinhibitionofNADPHoxidasesin vascularsmoothmusclecellsexposedtoangiotensinII[37].How- ever,astheH2O2-inducedRONSgenerationincardiacmyocytes cannotbefullyrecoveredinthepresenceofbothPPAR␤/␦ago- nistandantagonist(Fig.4),wecannotexcludethepossibilitythat

Fig.7. PPAR␤/␦activationinhibitsMMP-2andMMP-9geneexpression.Cardiacmyocyteswereeitherleftuntreatedortheywerepre-treatedwithPPAR␤/␦agonistGW0742 (GW,50nM)and/ortheantagonistGSK0660(GSK,0.5␮M)for10min.ThentheywereincubatedintheabsenceorpresenceofH2O2(0.1mM)for3h.(A,B)TotalRNAwas extractedandMMP-2andMMP-9weredeterminedbyreal-timeRT-PCR.LevelsofmRNAwerenormalizedtothemRNAlevelsofthehousekeepinggene␤-actin.Results aremean±SEMof4–5experimentswithindependentcellpreparations.*p<0.05comparedwithcontrol,#p<0.05comparedwithcellstreatedonlywithH2O2,ˆp<0.05 comparedwithcellstreatedwithH2O2andPPAR␤/␦agonistGW0742.

(8)

E.Barlakaetal./PharmacologicalResearch95–96(2015)102–110 109

theagonistGW0742hasanadditionalanti-oxidanteffectindepen- dentofPPAR␤/␦activation.Infact,wehavepreviouslyshownthat PPAR␤/␦agonistGW0742caninhibitthephenylephrine-induced RONS generation in cardiac myocytes through a non-genomic redoxmechanism[38].

Bcl-2 family proteins are key components of the apoptotic processincardiacmyocytes[5].Bcl-2proteinscanberegulated eitherbytranscription, heterodimerizationor proteolyticcleav- age,whereastheirphosphorylationstatusplaysimportantrolein thoseevents.TreatmentwithPPAR␤/␦agonistGW0742reversed thedetrimentaleffectofH2O2onBcl-2expressionwhileitledtoa significantdecreaseoftheproapoptoticBaxproteinlevels(Fig.6).

These resultsare consistentwith previousstudies demonstrat- ingcardioprotectionassociatedwithmodulationofcardiaclevels ofantiapoptotic(Bcl-2andBcl-xl)orproapoptotic(BaxandBad) proteins[17,31,39].Thepresent datado not documenta direct causallinkbetweenPPAR␤/␦activationandmodulationofBcl-2 orBaxexpression.ItispossiblethattheobservedBcl-2upregula- tioninourexperimentalsettingresultsfromthePPAR␤/␦-induced ameliorationofoxidativestressorPPAR␤/␦crosstalkwithother signalingpathways. Furthermore,wecannot excludethepossi- bility that besides Bcl-2 and Bax, other proteins acting in the mitochondrial-dependentapoptoticpathwayarealsoaffectedby PPAR␤/␦activationandmightcontributetotheobservedcardio- protectiveeffects.

Increased RONS promote MMP activities. MMP activation is associatedwithdegradationandremodelingofextracellularmatrix underpathologicalconditions,thusleadingtoheartfailure[40].

It is now becoming clear that, beyond their effects on matrix metabolism,MMPs(inparticularMMP-2)arecapableofmodulat- inginflammatorypathwaysbyprocessingcytokines,chemokines, and growth factors [41] as well as affecting many other rele- vantmediatorsinvolvedinavarietyoffunctionsandpathogenic mechanismsleadingtocardiacdysfunction[30,42].Infact,MMP- 2hasbeenshowntobeinvolvedintheintracellulardegradation of troponinI [43], myosinlight chain 1 [44] and nuclearPARP [45,46].Inhibition of MMP activityhas been shown to protect themyocardiumformoxidative/nitrosativestress-inducedinjury suggestingnoveltargetsforcardiacfailuremanagement[10,47].

ThedataofthisstudyclearlydemonstratethatPPAR␤/␦agonist GW0742 downregulates the H2O2-induced MMP-2and MMP-9 geneoverexpression(Fig.7)incardiacmyocytes.Moreoverthis effect is reversed by administration of the PPAR␤/␦ antagonist GSK0660.ItisnotclearwhetherthereisadirectlinkbetweenPPAR activationandMMPinhibition.TheinhibitoryeffectofPPARson NF-␬Biswelldocumented[19],whereastherearereportsdemon- stratingaroleofNF-␬BintheactivationofMMPs[48,49].Thus, the observed MMP dowregulation in cardiac myocytes (Fig.7) maybetheresultoftheanti-inflammatoryactionofPPAR␤/␦.In thiscontext,ithasbeenshownthataPPAR␤/␦agonistmarkedly suppressestheIL-1␤-inducedMMP-2and MMP-9expressionin vascular smooth muscle cells [50]. This issue warrants further investigation.Furthermore,wecannotruleoutthepossibilitythat MMP suppressionby the agonistGW0742 in our experimental model might be a secondary effect through themodulation of oxidativestress.

Inconclusion,ourresultsdemonstratethatthePPAR␤/␦ago- nistGW0742inhibitscardiacmyocyteinjurybyreducingoxidative stressandinhibitingMMPexpressionand apoptosis.Itsprotec- tiveeffectsaredependentontheactivationofPPAR␤/␦because theyareabolishedbypretreatmentwiththePPAR␤/␦antagonist GSK0660.Althoughadditionalexperimentsareneededtoclarify theroleofPPAR␤/␦incardiacmyocytesapoptosis,theresultspre- sentedheresuggestthattheconnectionbetweenPPARsignaling andMMPsuppressionmightrepresentanewtargetfortheman- agementofoxidativestress-inducedcardiacdysfunction.

Acknowledgements

Theworkhasbeenco-financedbytheEuropeanUnion(Euro- peanSocialFund)andGreekNationalFunds(GSRT-HUN57),and the Hungarian Scientific Research Fund: OTKA PD 106001. G.A holdsa “JánosBolyai Fellowship”fromtheHungarian Academy of Sciences, P.J.holds a “Apáczai-Csere János Fellowship” from TÁMOP-4.2.4.A/2-11/1-2012-0001(NationalExcellenceProgram).

P.F.isaSzentágothaiFellowoftheNationalProgramofExcellence (TAMOP4.2.4.A/2-11-1-2012-0001).

References

[1]C.Ceconi,A.Boraso,A.Cargnoni,R.Ferrari,Oxidativestressincardiovascu- lardisease:mythorfact?Arch.Biochem.Biophys.420(2)(2003)217–221, http://dx.doi.org/10.1016/j.abb.2003.06.002.

[2]E. Takimoto, D.A. Kass, Role of oxidative stress in cardiac hypertrophy and remodeling, Hypertension 49(2) (2007) 241–248, http://dx.doi.org/

10.1161/01.HYP.0000254415.31362.a7.

[3]R.S.Whelan,V.Kaplinskiy,R.N.Kitsis,Celldeathinthepathogenesisofheart disease:mechanismsandsignificance,Annu.Rev.Physiol.72(2010)19–44, http://dx.doi.org/10.1146/annurev.physiol.010908.163111.

[4]A.Clerk,S.M.Cole,T.E.Cullingford,J.G.Harrison,M.Jormakka,etal.,Regula- tionofcardiacmyocytecelldeath,Pharmacol.Ther.97(3)(2003)223–261, http://dx.doi.org/10.1016/S0163-7258(02)00339-X.

[5]A.B.Gustafsson,R.A.Gottlieb,Bcl-2familymembersandapoptosis,takento heart,Am.J.Physiol.CellPhysiol.292(1)(2007)C45–C51,http://dx.doi.org/

10.1152/ajpcell.00229.2006.

[6]S.N.Willis,J.M.Adams,Lifeinthebalance:howBH3-onlyproteinsinduce apoptosis,Curr. Opin. CellBiol.17(6) (2005)617–625,http://dx.doi.org/

10.1016/j.ceb.2005.10.001.

[7]A.Page-McCaw,A.J.Ewald,Z.Werb,Matrixmetalloproteinasesandthereg- ulationoftissueremodelling,Nat.Rev.Mol.CellBiol.8(3)(2007)221–233, http://dx.doi.org/10.1038/nrm2125.

[8]D.A.Siwik,W.S.Colucci,Regulationofmatrixmetalloproteinasesbycytokines andreactiveoxygen/nitrogenspeciesinthemyocardium,HeartFail.Rev.9(1) (2004)43–51.

[9]J.Shen,D.O’Brien,Y.Xu,Matrixmetalloproteinase-2contributestotumor necrosisfactoralphainducedapoptosisinculturedratcardiacmyocytes, Biochem.Biophys.Res.Commun.347(4)(2006)1011–1020,http://dx.doi.org/

10.1016/j.bbrc.2006.07.002.

[10]G.Dormán,S.Cseh,I.Hajdú,L.Barna,D.Kónya,etal.,Matrixmetalloproteinase inhibitors:acriticalappraisalofdesignprinciplesandproposedtherapeu- ticutility, Drugs70 (May(8))(2010) 949–964,http://dx.doi.org/10.2165/

11318390-000000000-00000.

[11]Q.Yang,Y. Li,Roles ofPPARsonregulatingmyocardialenergyand lipid homeostasis,J.Mol.Med.(Berl.)85(7)(2007)697–706,http://dx.doi.org/

10.1007/s00109-007-0170-9.

[12]G.D.Girnun,F.E.Domann,S.A.Moore,M.E.Robbins,Identificationofafunc- tionalperoxisomeproliferator-activatedreceptorresponseelementintherat catalasepromoter,Mol.Endocrinol.16(12)(2002)2793–2801,http://dx.doi.

org/10.1210/me.2002-0020.

[13]Q.Ding,T.Jin,Z.Wang,Y.Chen,Catalasepotentiatesretinoicacid-induced THP-1monocytedifferentiationintomacrophagethroughinhibitionofper- oxisomeproliferator-activatedreceptorgamma,J.Leukoc.Biol.81(6)(2007) 1568–1576,http://dx.doi.org/10.1189/jlb.1106672.

[14]T.Ravingerova,A.Adameova,S.Carnicka,M.Nemcekova,T.Kelly,J.Matejikova, E.Barlaka,E.A.Lazou,etal.,TheroleofPPARinmyocardialresponsetoischemia innormalanddiseasedheart,Gen.Physiol.Biophys.30(4)(2011)329–341, http://dx.doi.org/10.4149/gpb201104329.

[15]C.Lotz,M.Lazariotto, A.Redel,T.M.Smul,J. Stumpner,etal.,Activation ofperoxisome-proliferator-activated receptors and mediatesremote ischemicpreconditioningagainstmyocardialinfarctioninvivo,Exp.Biol.Med.

236(1)(2011)113–122,http://dx.doi.org/10.1258/ebm.2010.010210.

[16]T.Ravingerová,S.Carnická,M.Nemˇceková,V.Ledvényiová,A.Adameová, T. Kelly, E. Barlaka, E. Galatou, V.K. Khandelwal, A. Lazou, PPAR-alpha activationas a preconditioning-like intervention in rats in vivo confers myocardialprotectionagainstacuteischaemia-reperfusioninjury:involve- ment ofPI3K-Akt, Can. J. Physiol. Pharmacol. 90 (8) (2012) 1135–1144, http://dx.doi.org/10.1139/y2012-052.

[17]E.Barlaka,V.Ledvényiová,E.Galatou,M.Ferko,S. ˇCarnická,etal.,Delayed cardioprotectiveeffectsofWY-14643areassociatedwithinhibitionofMMP- 2andmodulationofBcl-2familyproteinsthroughPPAR-␣activationinrat heartssubjectedtoglobalischaemia-reperfusion,Can.J.Physiol.Pharmacol.

91(8)(2013)608–616,http://dx.doi.org/10.1139/cjpp-2012-0412.

[18]I. Inoue, S. Goto, T. Matsunaga,T. Nakajima, T. Awata, et al., The lig- ands/activatorsforperoxisomeproliferator-activatedreceptoralpha(PPARal- pha)andPPARgammaincreaseCu2+,Zn2+-superoxidedismutaseanddecrease p22phoxmessageexpressionsinprimaryendothelialcells,Metabolism50(1) (2001)3–11,http://dx.doi.org/10.1053/meta.2001.19415.

[19]P.J. Smeets, A. Planavila, G.J. vander Vusse, M.van Bilsen, Peroxisome proliferator-activated receptors and inflammation:take itto heart,Acta

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

Panels a–c compare the distributions of the disorder levels, functional similarity (BP SemSim), and the mRNA co-expression levels, respectively, of interaction partners in the

Similar to the previous models, differences in mRNA expression levels of angio- genic markers Hif1α and Vegfα were not observed and body weights showed no significant differences

In order to verify the microarray data and further examine the role of histone acetylation during the UVB response of the chromosome 11 MMP gene cluster, we determined the mRNA

Adenocarcinoma showed almost no expression of GST and signifi cantly higher levels of MMP-9 than Barrett and concomitant dysplasia.. Alterations of GST and MMP-9 were

andrei lumbricin (Lumbr) and LuRP mRNA expression levels 2. from various tissues of

Major research areas of the Faculty include museums as new places for adult learning, development of the profession of adult educators, second chance schooling, guidance

The decision on which direction to take lies entirely on the researcher, though it may be strongly influenced by the other components of the research project, such as the

Therefore, we designed studies in which preconditioning was induced by rapid cardiac pacing and changes in mRNA expression of 29 genes were analyzed immediately, 6, 12, and 24 h