ebook img

Four-and-a-half LIM domains proteins are novel regulators of the protein kinase D pathway in cardiac myocytes. PDF

1.3 MB·English
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 Four-and-a-half LIM domains proteins are novel regulators of the protein kinase D pathway in cardiac myocytes.

Biochem.J.(2014)457,451–461(PrintedinGreatBritain) doi:10.1042/BJ20131026 451 Four-and-a-half LIM domains proteins are novel regulators of the protein kinase D pathway in cardiac myocytes KonstantinaSTATHOPOULOU*†,FriederikeCUELLO*†,AlexandraJ.CANDASAMY*,ElizabethM.KEMP*,ElisabethEHLER*, g RobertS.HAWORTH*andMetinAVKIRAN*1 r *CardiovascularDivision,King’sCollegeLondonBritishHeartFoundationCentre,LondonSE17EH,U.K. o †InstituteofExperimentalPharmacologyandToxicology,UniversityMedicalCenterHamburg-Eppendorf,Martinistr.52,Hamburg20146,Germany . j m e PKD (protein kinase D) is a serine/threonine kinase implicated response to endothelin 1, but not to the α -adrenoceptor 1 h in multiple cardiac roles, including the phosphorylation of the agonistphenylephrine.Incontrast,selectiveknockdownofFHL2 classIIHDAC5(histonedeacetylaseisoform5)andtherebyde- expression caused a significant reduction in PKD activation c repression of MEF2 (myocyte enhancer factor 2) transcription and HDAC5 phosphorylation in response to both stimuli. o factoractivity.InthepresentstudyweidentifyFHL1(four-and- Interestingly, neither intervention affected MEF2 activation by a-halfLIMdomainsprotein1)andFHL2asnovelbindingpartners endothelin 1 or phenylephrine. We conclude that FHL1 and i forPKDincardiacmyocytes.Thiswasconfirmedbypull-down FHL2 are novel cardiac PKD partners, which differentially b assaysusingrecombinantGST-fusedproteinsandheterologously facilitatePKDactivationandHDAC5phosphorylationbydistinct . orendogenouslyexpressedPKDinadultratventricularmyocytes neurohormonalstimuli,butareunlikelytoregulateMEF2-driven w orNRVMs(neonatalratventricularmyocytes)respectively,and transcriptionalreprogramming. byco-immunoprecipitationofFHL1andFHL2withGFP–PKD1 w fusion protein expressed in NRVMs. In vitro kinase assays showed that neither FHL1 nor FHL2 is a PKD1 substrate. Keywords:cardiacmyocyte,four-and-a-halfLIM(FHL),histone w SelectiveknockdownofFHL1expressioninNRVMssignificantly deacetylase, neurohormonal stimulation, protein kinase, signal inhibited PKD activation and HDAC5 phosphorylation in transduction. l a INTRODUCTION The functional significance of PKD1in cardiac myocyte n (patho)physiology has recently started to be unveiled by both The PKD (protein kinase D) family of serine/threonine kinases in vitro and in vivo studies. We have shown previously that r consistsofthreemembers,PKD1,PKD2andPKD3,andbelongs PKD1mayregulatecardiacmyofilamentfunctionandtheCa2+ u to the CaMK (Ca2+/calmodulin-dependent protein kinase) sensitivity of contraction by phosphorylating cTnI (inhibitory o superfamily. These PKD isoforms share the common structural subunit of cardiac troponin) at Ser22/Ser23 [7,8] and cMyBP-C features of a C-terminal catalytic domain and an N-terminal (cardiac myosin-binding protein C) at Ser302 [9]. Furthermore, J regulatory domain. Components of the regulatory domain PKD1 has been proposed to facilitate cardiac hypertrophy autoinhibit the activity of the catalytic domain in unstimulated through the phosphorylation of HDAC5 (histone deacetylase l a cells and promote PKD association with the plasma and isoform 5) at Ser259 and Ser498 [10]. Nuclear HDAC5 associates intracellularmembranesafterstimulationwithhormones,growth with and represses the activity of MEF2 (myocyte enhancer c factors,neurotransmitters,chemokinesandbioactivelipids[1,2]. factor 2) transcription factors, which drive the transcriptional i In cardiac myocytes, the most abundantly expressed PKD reprogrammingthatprecipitatespathologicalcardiachypertrophy m family member is PKD1, which is activated after stimulation andremodelling.Inresponsetopro-hypertrophicneurohormonal of diverse GPCRs (G-protein-coupled receptors) that signal via stimuli, activated PKD1 phosphorylates HDAC5 at Ser259 and e Gα ,includingα -adrenergic,ET1(endothelin1)andangiotensin Ser498, thus inducing the binding of 14-3-3 proteins to these q 1 h II receptors [3–5]. The principal PKD activation mechanism sitesandrevealingaNES(nuclearexportsequence)thattriggers involves recruitment of the kinase to plasma or intracellular HDAC5 extrusion from the nucleus to the cytosol, through c membranesbyDAG(diacylglycerol)andtransphosphorylationof a mechanism that is mediated by the CRM1 (chromosome o itsactivationloopataminoacidresiduesSer744andSer748(amino region maintenance 1) protein [10,11]. HDAC5 nuclear export acidnumberingreferstomurinePKD1)byactivatednovelPKC de-represses MEF2 transcriptional activity, which then drives i (protein kinase C) isoforms. The resulting PKD activation then pro-hypertrophic gene expression [12–14]. Studies in mice B leads to both autophosphorylation at residue Ser916 and trans- with cardiac-specific deletion [15] or overexpression [16] of phosphorylation of PKD substrates, which include transcription PKD1 corroborate a key role for PKD1in pathological cardiac factors,proteinsinvolvedincellmotilityandvesiclefissionfrom remodelling, and PKD1 expression and activation have been theGolgiapparatus,otherkinasesandsarcomericproteins[1,2,6]. showntobeincreasedinfailinghumanmyocardium[17]. Abbreviations: ARVM, adult rat ventricular myocyte; BPKDi, bipyridyl PKD inhibitor; CaMK, Ca2+/calmodulin-dependent protein kinase; caPKD, constitutivelyactivecatalyticdomainofPKD;cMyBP-C,cardiacmyosin-bindingproteinC;CRM1,chromosomeregionmaintenance1;cTnI,inhibitory subunitofcardiactroponin;ERK,extracellular-signal-regulatedkinase;ET1,endothelin1;FHL,four-and-a-halfLIMdomains;HDAC,histonedeacetylase; IVK,invitrokinase;MEF2,myocyteenhancerfactor2;MOI,multiplicityofinfection;MuRF,muscleRINGfinger;NRVM,neonatalratventricularmyocyte; PE,phenylephrine;pfu,plaque-formingunit;PKC,proteinkinaseC;PKD,proteinkinaseD;TAC,transverseaorticconstriction. 1 Towhomcorrespondenceshouldbeaddressed([email protected]). © 2014 The Author(s) (cid:2)c TheAuthorsJournalcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. 452 K.Stathopoulouandothers The key roles proposed for PKD activity in cardiac with an adenoviral vector expressing murine PKD1 at a MOI (patho)physiologymakeimprovedunderstandingofthemolecu- (multiplicityofinfection)of100pfu(plaque-formingunit)/cell. larmechanismsunderlyingboththeupstreamregulationandthe ARVMs were maintained in culture for 24h before use in downstream actions of this kinase in the heart an imperative. experiments. Towards this objective, in a previous study [7], we performed a yeast two-hybrid screen of a human cardiac cDNA library, whichidentifiedFHL2(four-and-a-halfLIMdomainsprotein2) IsolationandcultureofNRVMs(neonatalratventricularmyocytes) asanovelbindingpartnerforthePKD1catalyticdomain.Inthe NRVMs were isolated and maintained in culture as described presentstudy,wehaveconfirmedandcharacterizedtheinteraction previously[8],andtransferredtomaintenancemedium[DMEM offull-lengthPKD1withFHL2aswellasthehighlyhomologous (Dulbecco’s modified Eagle’s medium)/M199 4:1 (v/v), 100 FHLisoformFHL1(bothofwhichareabundantlyexpressedin i.u./ml penicillin and streptomycin] 24h after isolation. For the heart [18]) in cardiac myocytes and explored the potential RNAiexperiments,themaintenancemediumuseddidnotcontain functionalsignificanceoftheseFHLisoformsinregulatingPKD antibiotics. activityanddownstreamactionsinthatcelltype. ExpressionofrecombinantFHLproteins EXPERIMENTAL PlasmidsencodingGST–FHL1andGST–FHL2fusionproteins Materials were transformed into Escherichia coli BL21 (DE3) pLysS cells (Invitrogen). Cells were grown at 37◦C in LB medium Rabbit polyclonal antibodies against phosphorylated supplemented with 100μg/μl ampicillin and 100μM ZnCl . (pSer744/Ser748 and pSer916) PKD were from Cell Signaling 2 When D reached 0.6–0.8, cells were induced to express the Technology.RabbitpolyclonalantibodiesagainsttotalPKDwere 600 GST-fused proteins for 3h by adding IPTG to 1mM. Cells fromCellSignalingTechnologyandSantaCruzBiotechnology. were then centrifuged at 5000g for 5min (4◦C) in a Sorvall A rabbit polyclonal antibody against phosphorylated (pSer498) RC5Bcentrifuge(GSArotor),pelletswereresuspendedinPBS HDAC5 and a mouse monoclonal antibody against FHL1 were containing 1% (v/v) Triton X-100 and CompleteTM protease fromAbcam.AmousemonoclonalantibodyagainstFHL2was inhibitors (Roche) and suspensions were then sonicated on ice from Medical and Biological Laboratories. Mouse monoclonal (threetimesfor15s,SonicsMaterialsUltrasonicProcessor).For antibodiesagainstGFPandGSTwerefromRocheandSantaCruz thepurificationofsolublerecombinantGSTandGST–FHL1,the Biotechnologyrespectively.Expressionvectorsforexpressionof lysatewasclearedbycentrifugationat12000gfor30min(4◦C) recombinantmouseFHL1andhumanFHL2proteinsweregifts (rotorSS34)andtheproteinwaspurifiedbypassingthecleared from Stephan Lange (Department of Medicine, UC San Diego, lysate through a glutathione–Sepharose 4B prepacked column CA,U.S.A.)[19].RecombinantPKDcatalyticdomainexpressed (GE Healthcare) according to the manufacturer’s instructions. in Sf21 insect cells was a gift from Harold Jeffries and Peter J. For the purification of insoluble recombinant GST–FHL2, the Parker (London Research Institute, Cancer Research UK) and lysatewascentrifugedat20000gfor30min(4◦C)inaBeckman recombinant human TnI was a gift from Douglas G. Ward and L60 ultracentrifuge (rotor SW 41 Ti). The pellet was then Ian Trayer (School of Biosciences, University of Birmingham, resuspended in PBS containing 1% (v/v) Triton X-100 and Birmingham, U.K.) [7]. BPKDi (bipyridyl PKD inhibitor) was protease inhibitors and centrifuged again, as described above. custom-synthesized by the MRC Protein Phosphorylation Unit, The pellet was then dissolved in PBS containing 1% (v/v) UniversityofDundee,Dundee,U.K.[20].Theadenoviralvectors Triton X-100, protease inhibitors and 8M urea and centrifuged used to express mouse PKD1 (AdV:wtPKD1) and enhanced at20000gfor15min(4◦C).Thesupernatant,whichcontained GFP (AdV:GFP) were prepared as described previously [8]. thedenaturedrecombinantGST–FHL2,wasdialysedinrefolding The adenoviral vector encoding human PKD1 tagged at the buffercontaining50mMTris/HCl,pH8.0,50mMNaCl,1mM N-terminus with GFP (AdV:GFP–PKD1) was a gift from Jody DTT and 0.1mM ZnCl . GST–FHL2 was purified using a Martin (Department of Cell and Molecular Physiology, Loyola 2 glutathione–Sepharose4Bprepackedcolumn,asnotedabove. University Chicago, IL, U.S.A.) [17]. The MEF2-luciferase reporter construct was a gift from Eric Olson (Department of Molecular Biology, UT Southwestern, TX, U.S.A.) [14] and GSTpull-downassay the promoter from this (comprising three tandem repeats of the MEF2-bindingsitefromthedesminenhancer)wassubclonedinto For the GST pull-down assay, lysates from ARVMs (treated apGL4.24vectorencodingthesyntheticluciferasereportergene with vehicle or 100nM PMA for 10min) or NRVMs (treated luc2P(Promega).ThevectorencodingRenillaluciferasereporter with vehicle or 10nM ET1 for 10min) prepared in lysis (pRL-null)wasfromPromega.ET1wasfromCalbiochem.PMA buffer [containing 50mM Tris/HCl, pH7.4, 0.05mM ZnCl2, and PE (phenylephrine) were from Sigma–Aldrich. Secondary 2mM DTT and 1% (v/v) Triton X-100] were clarified by antibodies and ECL kits were from GE Healthcare. All other centrifugation (25000g, 20min, 4◦C) in an Eppendorf 5417R chemicals were from Sigma–Aldrich, Life Technologies or centrifuge.Eachsupernatantwaspre-clearedbyincubationwith FisherScientific,unlessotherwisestated.Theinvestigationwas glutathione–Sepharose 4B beads (GE Healthcare) for 1h under performed in accordance with the Home Office Guidance on rotation(4◦C),thensplitinequalvolumesandincubatedfor2h the Operation of the Animals (Scientific Procedures) Act 1986 under rotation (4◦C) with 5μg of recombinant proteins (GST– (U.K.). FHL1,GST–FHL2andGST)pre-boundfor1htoglutathione– Sepharose 4B beads (4◦C). The complexes were pelleted by centrifugation(1000g,1min,4◦C),washedthreetimesinlysis buffer, resuspended in 2× SDS/PAGE sample buffer [125mM IsolationandcultureofARVMs(adultratventricularmyocytes) Tris/HCl,pH6.8,4%(w/v)SDS,20%(v/v)glycerol,6%(v/v) ARVMs were isolated and maintained in culture as described 2-mercaptoethanol and 0.02% Bromophenol Blue], boiled and previously [8]. Attached cardiac myocytes were transduced storedat −20◦Cuntiluse. ©(cid:2)c 20T1h4e TAhueth oArusthJoour(rsn)alcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. FHL1andFHL2arenovelregulatorsofPKD 453 Immunoprecipitation Luciferaseassays Onedayafterisolation,NRVMsweretransducedwithadenoviral NRVMsweretransfectedinitiallywithactiveorscrambledsiRNA vectorsexpressingeitherGFP–PKD1(atMOIof50pfu/cell)or sequences,asdescribedabove,and24hlaterwithacombination GFP (at MOI of 10 pfu/cell). After 24h, NRVMs were treated of0.6μgofMEF2-luc2Pluciferasereporterconstructand0.4μg for 20min with either ET1 (10nM) or vehicle and GFP–PKD1 of Renilla luciferase reporter construct, the latter using Escort or GFP was immunoprecipitated by using the GFP-Trap®_A III transfection reagent (Sigma). The medium was changed kit (Chromotek), according to the manufacturer’s instructions. to transfection medium without serum 5h later and the cells Briefly, NRVMs were lysed in a buffer containing 10mM incubatedforafurther3h,beforetheyweretreatedwithvehicle, Tris, pH7.5, 150mM NaCl and 0.5mM EDTA, supplemented ET1 (10nM) or PE (3μM, in the presence of 1μM atenolol) with 0.5% Nonidet P40 and protease inhibitors. Lysates were for 18–24h. Luciferase assays were performed using the Dual- incubated on ice for 30min and subsequently centrifuged for Glo®LuciferaseassaysystemandaGloMax®20/20luminometer 10minat20000g(4◦C).Theclearedlysateswerethendiluted (Promega), according to the manufacturer’s instructions. In to a final volume of 1ml with dilution buffer (containing experiments where BPKDi was used, NRVMs were processed 10mM Tris/HCl, pH7.5, 150mM NaCl, 0.5mM EDTA and in a similar manner, but without the siRNA transfection step; supplemented with protease inhibitors) and incubated under BPKDi(3μM)orvehiclewasadded30minbeforetreatmentwith rotation for 2h (4◦C) with GFP-Trap®_A beads. Beads were vehicle, ET1 or PE. Firefly luciferase activity was normalized thenwashedthreetimesindilutionbuffer,resuspendedin100μl against Renilla luciferase activity to account for transfection of 2× SDS/PAGE sample buffer, boiled and stored at −20◦C efficiency.DatawereexpressedrelativetothescrambledsiRNA- untiluse. control group (in experiments where FHL protein levels were knocked down by RNAi) or to the vehicle-control group (in experimentswherePKDactivitywaspharmacologicallyinhibited Invitrophosphorylationassay withBPKDi). Theinvitrophosphorylationassaywascarriedoutasdescribed previously[7].RecombinantGST,GST–FHL1,GST–FHL2and Immunoblotanalysis cTnI,aloneorincombination(200pmoleach)asindicated,were incubatedfor30minat37◦C,intheabsenceorpresenceofactive Immunoblotanalysiswasperformedasdescribedpreviously[7], PKD(35ng)togetherwith100μMATP,supplementedwith[γ- using specific antibodies for total or phosphorylated proteins as 32P]ATPinkinaseassaybuffer(30mMTris/HCl,pH7.4,15mM indicated. Specific protein bands were detected by ECL (GE MgCl ,1mMDTTand0.05mMZnCl ).Proteinswereresolved Healthcare) and phosphorylation status was quantified using 2 2 on 12% acrylamide gels, which were subsequently dried and a calibrated densitometer (GS-800TM, Bio-Rad Laboratories). subjectedtoautoradiography. Data were normalized to total protein loading (measured by densitometryintherespectiveimmunoblotsafterCoomassieBlue staining) and expressed relative to an internal standard sample RNAi includedinaseparatelane,topermitconsolidationofexperiments analysedondifferentgels. Synthetic siRNA sequences (denoted as ‘active’ siRNAs) and theirrespectivescrambledcounterparts(denotedas‘scrambled’ siRNAs) with 3(cid:3) dTdT overhangs were purchased from Life Statisticalanalysis Technologies.Thesequencesusedwere:5(cid:3)-GCCUGAAGUGCU- UUGACAA-3(cid:3) (active siRNA sequence for FHL1), 5(cid:3)-GCCA- Statistical comparisons were performed by one-way ANOVA AUGCGUUGUAUGCAA-3(cid:3) (scrambled siRNA sequence for followed by the Newman–Keuls post-hoc test. Quantitative FHL1), 5(cid:3)-GCAAGGACUUGUCCUACAA-3(cid:3) (active siRNA data are given as means+−S.E.M. and P<0.05 was considered sequence for FHL2) and 5(cid:3)-GCAUCAGCUGUAUCGACAA-3(cid:3) significant. (scrambledsiRNAsequenceforFHL2).NRVMsweretransfected with 40 nmol of active or scrambled siRNA sequences using RESULTS Lipofectamine RNAimax (Life Technologies), 24h after they were transferred to maintenance medium. The cells were then PKDinteractswithbothFHL1andFHL2 incubatedfor48hintransfectionmediumcontaining22%(v/v) The potential interactions of PKD with FHL1 and FHL2 were M199,4%(v/v)horseserumand74%(v/v)modifiedDulbecco’s initiallyinvestigatedthroughpull-downassaysusingrecombinant balanced salt solution buffer (116mM NaCl, 1mM NaH PO , 2 4 GST–FHL1andGST–FHL2proteinsandcelllysatesfromAR- 0.8mMMgSO ,5.5mMglucose,32.1mMNaHCO and1.8mM 4 3 VMsthatheterologouslyexpressedmousePKD1followingaden- CaCl ,pH7.2),afterwhichtimetheywereserum-starvedfor3h 2 oviralgenetransfer.ARVMsweretreatedwithvehicleorPMA(to before treatment for 20min with vehicle, ET1 (10nM) or PE activatePKD)toascertainthepotentialimpactofPKDactivation (3μM,inthepresenceof1μMatenololadded10minbeforePE). Atenololwasusedtoinhibitanyβ-adrenergicreceptoractivation, statusonanyinteractionwithFHL1orFHL2.PKD1proteinwas pulleddownbybothGST–FHL1andGST–FHL2,butnotbyGST whichisknowntocounteractPKDactivation[20,21]. alone(Figure1A,firstpanel),indicatingaspecificinteractionwith bothFHL1andFHL2.Asexpected,PMAmarkedlyincreasedthe phosphorylationstatusofPKD1atSer744/Ser748andSer916,reflect- PharmacologicalinhibitionofPKDactivity ingactivationofthekinase.PhosphorylatedPKD1associatedwith IntheexperimentswheretheselectivePKDinhibitorBPKDiwas bothFHL1andFHL2inthispull-downassay(Figure1A,second used,NRVMs were processedas describedabovefor theRNAi andthirdpanels),indicatingthatthePKD1–FHL1/2association experiments,butwithoutthetransfectionstep.BPKDi(3μM)or occursindependentlyofthePKD1activationstatus.Baitprotein vehiclewasadded30minbeforetreatmentwithvehicle,ET1or (GST–FHL1, GST–FHL2 or GST) content was comparable in PE. controlandstimulatedgroups(Figure1A,fourthandfifthpanels). © 2014 The Author(s) (cid:2)c TheAuthorsJournalcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. 454 K.Stathopoulouandothers Figure 2 Co-immunoprecipitation of endogenous FHL1 and FHL2 with heterologouslyexpressedPKD1 NRVMs transduced with AdV:GFP–PKD1 or AdV:GFP were treated with vehicle (C) or ET1 (10nM) for 20min and protein complexes associated with GFP–PKD1 or GFP were immunoprecipitatedusingGFP-Trapmethodology.AssociationofGFP–PKD1orGFPwith endogenouslyexpressedFHL1andFHL2wasdetectedbyimmunoblot(IB)analysisofthe immunoprecipitates (IP) using specific antibodies against FHL1 or FHL2. Expression and immunoprecipitationofGFP–PKD1andGFPwereverifiedbyimmunoblotanalysisusingaGFP antibody,whereasactivationstatusofthePKD1componentofGFP–PKD1wasdeterminedby immunoblotanalysisusingaphospho-specificpSer916PKDantibody.Samplesofthelysates beforetheIP(input,10%)werealsoincludedintheimmunoblotanalyses.Dataarerepresentative ofthreeindependentexperiments. Figure1 Pull-downofheterologouslyexpressedorendogenousPKD1with immunoblot assessment of PKD autophosphorylation status at recombinantFHL1andFHL2 Ser916 (Figure2,thirdpanel).Immunoblotanalysiswithananti- GFP antibody confirmed comparable expression and equally (A)ARVMstransducedwithAdV:wtPKD1weretreatedwithvehicle(C)orPMA(100nM)for efficient immunoprecipitation of GFP–PKD1 and GFP in the 10min.LysateswereincubatedwithpurifiedGST–FHL1,GST–FHL2orGSTbaitproteinsbound toglutathione–SepharosebeadsandthePKDpull-downwasassessedbyimmunoblot(IB) differentstudygroups(Figure2,fourthandfifthpanels).Taken analysisusingtotalorphospho-specific(pSer916orpSer744/Ser748)PKDantibodies.Samples together, the above results provide complementary biochemical ofthelysatesbeforethepull-downassay(input,10%)werealsoincludedintheimmunoblot evidence that FHL1 and FHL2 are novel interaction partners analysesandareshowninseparatepanelsreflectingdifferentexposuretimes.Baitprotein for PKD1 and that this interaction occurs independently of the contentofthepull-downswasdeterminedbyimmunoblotanalysisusingaGSTantibody.(B) activationstatusofthekinase. Asin(A),butusinglysatesfromNRVMstreatedwithvehicle(C)orET1(10nM)for10min. Immunoblotsarerepresentativeofthreeindependentexperiments. FHL1andFHL2arenotphosphorylatedbyPKD ToexploretheinteractionofendogenousPKDwithFHL1and Ourpreviousstudiesthatbuiltonthediscoveryofnovelcardiac FHL2, we additionally performed GST pull-down assays using PKD1interactionpartnersthroughayeasttwo-hybridscreen[7] cell lysates from NRVMs, in which PKD1 is more abundantly have led to the identification of cTnI and cMyBP-C as PKD expressed than in ARVMs [3]. PKD1 was pulled down by substratesandthemappingandfunctionalanalysisofthepertinent GST–FHL1 and GST–FHL2, but not by GST alone, reflecting substratephosphorylationsites[7–9].Inananalogousapproach, an interaction between the endogenous kinase and both FHL1 weexploredwhetherFHL1andFHL2arealsoPKDsubstrates, andFHL2(Figure1B,firstpanel).Theseinteractionswerealso byperformingIVK(invitrokinase)assaysinwhichthecaPKD retainedfollowingtheactivationofendogenousPKD(Figure1B, (constitutively active catalytic domain of PKD) was incubated second and third panels), this time achieved by exposure to a with recombinant GST, GST–FHL1 or GST–FHL2 proteins in physiologicalstimulus,ET1.Asabove,baitprotein(GST–FHL1, the presence of [γ-32P]ATP. Autoradiographic analysis revealed GST–FHL2 or GST) content was comparable in control and nodetectablephosphateincorporationintoGST,GST–FHL1or stimulatedgroups(Figure1B,fourthandfifthpanels). GST–FHL-2, even though robust phosphorylation of cTnI was We next investigated whether the PKD1–FHL1/2 interaction readily detectable (Figure 3A). To explore whether FHL1 or occurs in the cellular environment of cardiac myocytes. To FHL2 might affect directly the catalytic activity of PKD, we address this, we expressed GFP–PKD1 or GFP in NRVMs by also performed IVK reactions in which caPKD was incubated adenoviralgenetransferandemployedGFP-Trapmethodologyto with cTnI in the absence or presence of each of the GST-fused efficientlyimmunoprecipitateproteincomplexesassociatedwith proteins. These experiments revealed that cTnI phosphorylation these proteins. Immunoblot analysis revealed that endogenous was not affected by the presence of GST, GST–FHL1 or GST– FHL1andFHL2bothco-immunoprecipitatedwithGFP–PKD1, FHL2(Figure3B).SuchfindingsindicatethatFHL1andFHL2 but not with GFP, under basal conditions and after stimulation are not PKD substrates, which is consistent with the fact that with ET1 (Figure 2, first and second panels). Activation of theaminoacidsequencesoftheseproteinsdonotcontainPKD the PKD1 component of the heterologously expressed GFP– substrate consensus phosphorylation motifs, as revealed by in PKD1 fusion protein in response to ET1 was verified by silicoanalysis(http://phospho.elm.eu.org).Theyalsosuggestthat ©(cid:2)c 20T1h4e TAhueth oArusthJoour(rsn)alcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. FHL1andFHL2arenovelregulatorsofPKD 455 Figure3 FHL1andFHL2arenotPKDsubstrates (A)AutoradiogramsofinvitrophosphorylationassaysusingrecombinantcaPKDandrecombinantGST,GST–FHL1,GST–FHL2orcTnIproteinassubstrate.(B)Assayswerealsoconductedusing cTnIassubstrate,intheabsenceorpresenceofadditionalrecombinantGST,GST–FHL1orGST–FHL2protein.(C)Histogramshowingquantitativedatafromexperimentsasillustratedin(B). Reactionswereperformedinthepresenceof[γ-32P]ATPandproteinswereresolvedbySDS/PAGE(12%gel).ProteinloadingwasconfirmedbyCoomassieBluestaining.Dataarerepresentativeof threeindependentexperiments,andthequantitativedatain(C)aremeans+−S.E.M.(n=3). interaction with FHL1 or FHL2 does not inhibit the catalytic As illustrated in Figure 4, both stimuli significantly increased activityofcaPKDtowardsanestablishedsubstrate,cTnI. PKD autophosphorylation at Ser916 and transphosphorylation at Ser744/Ser748 in control cells transfected with scrambled siRNA.KnockingdownFHL1expressionsignificantlyattenuated FHL1andFHL2facilitatePKDphosphorylationincardiacmyocytes the increase in PKD phosphorylation in response to ET1 To explore the potential roles of FHL1 and FHL2in regulating (Figure 4A), but had no significant effect on the increase in the activation and functions of endogenous PKD in cardiac PKDphosphorylationinresponsetoPE(Figure4B).Incontrast, myocytes, we applied a loss-of-function approach and knocked knockdown of FHL2 expression significantly attenuated the down FHL1 or FHL2 protein expression by RNAi through increases in PKD phosphorylation in response to both ET1 transfection of NRVMs with synthetic siRNA duplexes specific (Figure 4C) and PE (Figure 4D), with a greater inhibitory foreachFHLmRNAsequence.TransfectionwithFHL1siRNA effect on the latter. These findings suggest that FHL1 and caused an approximately 75% decrease in FHL1 protein FHL2 facilitate PKD activation by multiple neurohormonal expressionrelativetocontrolcellstransfectedwithascrambled stimuli,potentiallyinastimulus-dependentmanner,bypromoting siRNA sequence, with no apparent effect on FHL2 expression transphosphorylation of the PKD activation loop. Interestingly, (Supplementary Figures S1A and S1B at http://www.biochemj. theeffectsofsimultaneousknockdownofbothFHL1andFHL2 org/bj/457/bj4570451add.htm). Similarly, transfection with onPKDphosphorylationweresimilartotheeffectsofselective FHL2 siRNA caused a comparable decrease in FHL2 protein FHL2 knockdown, with no indication of an additive effect expression relative to cells transfected with a scrambled siRNA (Supplementary Figure S2 at http://www.biochemj.org/bj/457/ sequence, with no compensatory change in FHL1 expression bj4570451add.htm). (Supplementary Figures S1C and S1D). We then examined the FHL2hasbeenreportedtopromotethestabilityofmyocardin consequences of the selective knockdown of each FHL isoform andmyocardin-relatedtranscriptionfactor-Abyprotectingthese on PKD activation in response to stimulation with ET1 or PE. proteinsfromproteasome-mediateddegradation[22],raisingthe © 2014 The Author(s) (cid:2)c TheAuthorsJournalcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. 456 K.Stathopoulouandothers Figure4 EffectofFHL1orFHL2knockdownonET1-andPE-inducedphosphorylationofendogenousPKD NRVMsweretransfectedwitheitherscrambledsiRNAoractivesiRNAduplexestargetedatFHL1(AandB)orFHL2(CandD)transcripts.After48h,cellsweretreatedwithvehicle(C)orET1 (10nM)(AandC)orvehicle(C)orPE(3μM)(BandD)for20min.PhosphorylationstatusofendogenousPKDatSer916andSer744/Ser748wasdeterminedbyimmunoblot(IB)analysisusing appropriatephospho-specificantibodies.Proteinloadingwasconfirmedusingananti-PKDantibodyandbyCoomassieBluestaining.Individualimmunoblotsillustraterepresentativeexperiments, andhistogramsshowquantitativedataasmeans+−S.E.M.(n=7–8).*P<0.05. ©(cid:2)c 20T1h4e TAhueth oArusthJoour(rsn)alcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. FHL1andFHL2arenovelregulatorsofPKD 457 possibilitythat,ifFHL1orFHL2similarlyprotectedPKDprotein OnepossibleexplanationforthislackofeffectofFHLisoform from degradation, the apparent effects of FHL knockdown on knockdownonMEF2activation,despiteasignificantattenuation PKD phosphorylation described above might actually reflect a of PKD activation, is that under our experimental conditions decrease in total PKD protein levels. We therefore analysed PKDactivityisnotanimportantmediatorofMEF2activationin total PKD protein levels in samples after FHL1 or FHL2 responsetoET1orPE.TohelpconfirmtheroleofPKDactivity knockdown.Despiteatendency,therewasnosignificantreduction in MEF2 activation in NRVMs, therefore, we also investigated in PKD protein expression with knockdown of either FHL1 or the effects of the selective PKD inhibitor BPKDi [20,23] under FHL2(SupplementaryFigureS3athttp://www.biochemj.org/bj/ thesameexperimentalconditions.Consistentwithpreviouswork 457/bj4570451add.htm). Thus the negative effects of reduced [23],pre-treatmentofNRVMswithBPKDicompletelyabolished FHL isoform expression by RNAi on ET1- and PE- ET1-andPE-inducedincreasesinbothPKDautophosphorylation induced increases in PKD autophosphorylation at Ser916 and and HDAC5 phosphorylation (Figure 7A). Furthermore, pre- transphosphorylation at Ser744/Ser748 (Figure 4) most likely treatment with BPKDi significantly attenuated ET1- and PE- indicate an attenuation of PKD activation, rather than simply induced increases in MEF2 activity (Figures 7B and 7C). Thus reflectingreducedPKDproteinexpression. it appears that FHL1 and FHL2 facilitate PKD activation and HDAC5phosphorylation,butnotMEF2activationfollowingET1 andPEstimulation,despitethefactthatPKDactivityisindeedan FHL1andFHL2facilitateHDAC5phosphorylationincardiac importantmediatorofbothHDAC5phosphorylationandMEF2 myocytes activationundertheseexperimentalconditions. ToexplorewhethertheattenuationofPKDactivationisparalleled by reduced phosphorylation of a functionally important cellular PKD substrate, we also studied the effects of FHL1 and DISCUSSION FHL2 knockdown on the phosphorylation status of HDAC5 The present study provides novel information concerning the at Ser498, an established PKD substrate in cardiac myocytes regulation of PKD activity and functions in cardiac myocytes, [10,11].SelectiveFHLisoformknockdowninNRVMsattenuated throughthefollowing principalfindings:(i) PKDinteracts with HDAC5 phosphorylation in response to stimulation with ET1 two members of the FHL protein family, FHL1 and FHL2; (ii) or PE (Figure 5), in a manner that mirrored the effects of FHL1andFHL2arenotPKDsubstratesanddonotinhibitPKD these loss-of-function interventions on PKD phosphorylation catalytic activity; (iii) FHL1 and FHL2 differentially regulate (Figure 4). Thus selective knockdown of FHL1 expression PKDactivationafterneurohormonalstimulation;and(iv)FHL1 significantlyattenuatedtheincreaseinHDAC5phosphorylation and FHL2 do not regulate MEF2 activity after neurohormonal in response to ET1 compared with control cells transfected stimulation. with a scrambled siRNA sequence (Figure 5A), but had a non- FHL1andFHL2arenon-enzymaticproteinsthatareexpressed significant effect on the increase in HDAC5 phosphorylation in primarilyinstriatedmuscles,includingtheheart[18,24,25].Their response to PE (Figure 5B). In contrast, selective knockdown importancefornormalmusclefunctionisevidencedbythefact of FHL2 expression significantly attenuated the increases in thatmutationsinthegenesencodingtheseproteinsfhl1andfhl2, HDAC5 phosphorylation in response to both ET1 (Figure 5C) areassociatedwithvariousskeletalandcardiacmyopathies[25]. and PE (Figure 5D). Therefore it appears that, in response The multiple LIM domains comprising FHL1 and FHL2 form to neurohormonal stimulation in NRVMs, FHL1 and FHL2 a tandem zinc-finger structure that provides a modular protein- facilitate not only PKD activation, but also phosphorylation binding interface, through which FHL1 and FHL2 function as of the downstream PKD substrate HDAC5. Once again, the adaptors or scaffolds to support the assembly of multimeric effectsofsimultaneousknockdownofbothFHL1andFHL2on protein complexes and regulate the localization and activity of HDAC5phosphorylationweresimilartotheeffectsofselective their partners [24,25]. Examples include specific interactions FHL2 knockdown, with no indication of an additive effect betweenFHL1andFHL2withanarrayoftranscriptionfactors, (SupplementaryFigureS2). such as AP-1 (activator protein 1) [26] and HIF-1 (hypoxia- inducible factor 1) [27]. FHL proteins may co-activate or co- repress these transcription factors, depending on the identity FHL1andFHL2donotregulateMEF2activationincardiac of the interaction partner, the stimulus and the cell type [24– myocytes 27], thereby exerting regulatory effects on gene expression. PKD-mediated phosphorylation of HDAC5 at Ser498 is believed In addition, FHL1 and FHL2 have been reported to form to trigger HDAC5 nuclear export, with the consequent de- biologically significant interactions with the α5β1 and α7β1 repressionofMEF2activity[13,14].Wethereforehypothesized integrins [28,29], the muscle-specific RING finger proteins thatknockingdownFHL1orFHL2expressionmightalsohave MuRF1 and MuRF2 [30], the metabolic enzymes creatine an impact on ET1- and PE-induced MEF2 activation, as a kinase,adenylatekinaseandphosphofructokinase[19]andother consequenceofattenuatedPKDactivationandreducedHDAC5 proteinkinaseslikeERK2(extracellular-signal-regulatedkinase phosphorylation. To test this hypothesis, MEF2 transcriptional 2)[31,32].SuchinteractionsdefinethefunctionalrolesofFHL1 activity was monitored in NRVMs that were transfected andFHL2inregulatingbiologicalprocessessuchascelldivision, with a MEF2-luciferase reporter. As expected, stimulation of differentiation, migration and metabolism [24,25]. The present control NRVMs transfected with scrambled siRNA with either study adds PKD to the growing list of interaction partners for ET1 or PE significantly increased MEF2 activity (Figure 6). FHL1 and FHL2 and provides evidence that these interactions Surprisingly, however, transfection with active siRNA targeted areoffunctionalimportanceinregulatingPKDactivityincardiac at FHL1 (Figures 6A and 6B) or FHL2 (Figures 6C and 6D) myocytesinresponsetoneurohormonalstimulation. transcripts had no significant effect on ET1- and PE-induced PreviousstudieshaveexploredtherolesthatFHL1andFHL2 increases in MEF2 activity, despite again achieving marked may play in cardiac myocyte (patho)physiology through the reductionsinFHLproteinexpression(SupplementaryFigureS4 targeted disruption of fhl1 and fhl2 in mice. Such studies have athttp://www.biochemj.org/bj/457/bj4570451add.htm). revealedthatthegloballossofFHL1orFHL2proteindoesnot © 2014 The Author(s) (cid:2)c TheAuthorsJournalcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. 458 K.Stathopoulouandothers Figure5 EffectofFHL1orFHL2knockdownonET1-andPE-inducedphosphorylationofendogenousHDAC5atSer498 NRVMsweretransfectedwitheitherscrambledsiRNAoractivesiRNAduplexestargetedatFHL1(AandB)orFHL2(CandD)transcripts.After48h,cellsweretreatedwithvehicle(C)orET1(10nM) (AandC)orvehicle(C)orPE(3μM)(BandD)for20min.ThephosphorylationstatusofendogenousHDAC5wasdeterminedbyimmunoblot(IB)analysisusingaphospho-specificpSer498 HDAC5antibody.ProteinloadingwasconfirmedbyCoomassieBluestaining.Individualimmunoblotsillustraterepresentativeexperiments,andhistogramsshowquantitativedataasmeans+−S.E.M. (n=7–8).*P<0.05. impair cardiovascular development, or cause any spontaneous be isoform-specific and dependent on the nature of the stress cardiac phenotype [32–34]. They have also shown that the stimulusthattriggersthehypertrophicresponse.Interestingly,at targeted deletion of either fhl1 or fhl2 does not induce a leastinsomesettings,FHL1andFHL2appeartohavedifferential compensatory change in the cardiac expression of the other effects on cardiac myocyte ERK activity [31,32], raising the isoform [32,34], which is reminiscent of our observations possibilitythattheERKpathwaymaybecriticalinmediatingthe with individual knockdown of FHL1 or FHL2 expression in regulationofcardiachypertrophybyFHLproteins.Furthermore, NRVMs (Supplementary Figure S1). Nevertheless, when the recent evidence suggests a role for FHL2in the regulation of heartsofgene-targetedmiceweresubjectedtodifferenttypesof calcineurin/nuclear factor of activated T-cell signalling, another stress stimuli, some overt differences in the manifestation pro-hypertrophicpathway[35].FHL2repressescalcineurin,with of pathological phenotypes became apparent. For example, whichitco-localizesatthesarcomere[35].LossofFHL2from in comparison with their wild-type littermates, fhl1−/− mice thesarcomerehasbeenobservedinhumanheartfailuresamples, have been reported to develop diminished cardiac hypertrophy suggesting that changes in the subcellular distribution of the followingTAC(transverseaorticconstriction)orcardiac-specific proteinmaybeassociatedwithdiseasedevelopment[36]. Gα overexpression,suggestingapro-hypertrophicroleforFHL1 InourinvitrostudiesinNRVMs,selectiveknockdownofeither q [32].Onthecontrary,fhl2−/− micepresentedwithacomparable FHL1 or FHL2 inhibited PKD activation by pro-hypertrophic magnitude of cardiac hypertrophy relative to wild-type controls neurohormonal stimuli and attenuated the downstream phos- after TAC [34,35], but mounted an exaggerated hypertrophic phorylationofHDAC5,albeitwithsomedifferencesintheirim- response after chronic β-adrenergic receptor stimulation by pactontheresponsestoET1againstPE(discussedlater).Never- isoproterenolinfusion[33],suggestingananti-hypertrophicrole theless,MEF2activationbythesestimuliwasunaffectedbyFHL1 forFHL2inthelattersetting.ThustherolesthatFHL1andFHL2 or FHL2 knockdown (Figure 6). These data suggest that FHL playinregulatingcardiachypertrophyintheinvivosettingmay protein-mediated regulation of PKD activity is unlikely to play ©(cid:2)c 20T1h4e TAhueth oArusthJoour(rsn)alcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. FHL1andFHL2arenovelregulatorsofPKD 459 Figure6 EffectofFHL1orFHL2knockdownonET1-andPE-inducedactivationofendogenousMEF2 NRVMsweretransfectedwitheitherscrambledsiRNAoractivesiRNAduplexestargetedatFHL1(AandB)orFHL2(CandD)transcripts.After24h,cellswereco-transfectedwitha3×MEF2-firefly luciferasereportervectorandaRenillaluciferasecontrolvectorandtreatedwithvehicle(C)orET1(10nM)(AandC)orvehicle(C)orPE(3μM)(BandD)forafurther18–24h.Luciferaseactivity incelllysateswasassessedbyinvitroluminescenceassays,withluciferaseactivitynormalizedforRenillaluciferaseactivityineachsampletocorrectfortransfectionefficiency.Histogramsshow quantitativedataasmeans+−S.E.M.(n=4).*P<0.05. Figure7 EffectofpharmacologicalPKDinhibitiononET1-andPE-inducedHDAC5phosphorylationandMEF2activation (A)NRVMsweretreatedwithvehicle(C),ET1(10nM)orPE(3μM)for20min,followinga30-minpre-treatmentwithvehicleortheBPKDi(3μM).PKDautophosphorylationatSer916,totalPKD expressionandHDAC5phosphorylationatSer498wereassessedbyimmunoblot(IB)analysisusingappropriateantibodies,asindicated.MembraneswerestainedwithCoomassieBluetoconfirm equalproteinloading.Dataarerepresentativeofthreeindependentexperiments.(BandC)NRVMswereco-transfectedwitha3×MEF2-fireflyluciferasereportervectorandaRenillaluciferase controlvectorandtreatedwithvehicle(C),ET1(10nM)orPE(3μM)for18–24h,followinga30-minpre-treatmentwithvehicleortheBPKDi(3μM).Luciferaseactivityincelllysateswas assessedbyinvitroluminescenceassays,withluciferaseactivitynormalizedforRenillaluciferaseactivityineachsampletocorrectfortransfectionefficiency.Histogramsshowquantitativedataas means+−S.E.M.(n=4).*P<0.05. © 2014 The Author(s) (cid:2)c TheAuthorsJournalcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited. 460 K.Stathopoulouandothers acriticalroleincontrollingMEF2-drivencardiacmyocytetran- MEF2-driven transcriptional reprogramming towards cardiac scriptional reprogramming towards hypertrophy, despite its im- hypertrophy. pactonHDAC5phosphorylation.Inthisregard,althoughpharma- cologicalPKDinhibitionabolishedtheneurohormonalinduction ofPKDautophosphorylationandHDAC5transphosphorylation, AUTHORCONTRIBUTION itincompletely(albeitsignificantly)inhibitedMEF2activationin Metin Avkiran, Friederike Cuello, Elisabeth Ehler and Robert Haworth conceived and ourstudies(Figure7).Itispossible,therefore,thatacombination instigatedtheproject.KonstantinaStathopoulou,FriederikeCuelloandMetinAvkiran ofPKD-independentmechanisms,suchasthephosphorylationof planned the experiments, the majority of which were performed by Konstantina classIIHDACsbyCaMKII[37,38]orGRK5(G-protein-coupled StathopoulouwithcontributionsfromFriederikeCuello,AlexandraCandasamy,Elizabeth receptor kinase 5) [39], or their redox-mediated nuclear export KempandRobertHaworth.KonstantinaStathopoulouandMetinAvkirananalysedthedata andwrotethepaper. [20,40], or indeed HDAC-independent mechanisms mediated by p38-MAPK (mitogen-activated protein kinase) [41], ERK5 [42,43]ortheacetyltransferasep300[44],maybesufficienttoin- ACKNOWLEDGEMENTS ducesignificantMEF2activationandtherebyMEF2-driventran- scriptional reprogramming towards cardiac hypertrophy. In this WethankShineyRejiandUpamaliPerera(CardiovascularDivision,King’sCollegeLondon, context,itisalsoimportanttonotethatpharmacologicalinhibition London,U.K.)fortechnicalassistanceandStephanLange(DepartmentofMedicine,UC SanDiego,CA,U.S.A.)forkindlyprovidingthebacterialexpressionvectorsforGST–FHL1 ofPKDactivityhasbeenshownnottoattenuatecardiachyper- andGST–FHL2. trophyindifferentmodelsintheratinvivo[45],althoughcardiac- specificdeletionofPKD1inthemousesignificantlyinhibitedcar- diachypertrophyinresponsetoTACorchronicisoproterenolinfu- FUNDING sion[15].AplausibleinterpretationofsuchdataisthatPKD1reg- ulatescardiachypertrophythroughamechanismthatisindepend- ThisworkwassupportedbytheMedicalResearchCouncil[grantnumberG0800206]. entofitskinaseactivity,inwhichcaseFHL1-andFHL2-mediated regulationofneurohormonalPKDactivationwouldbemorelikely REFERENCES toregulateotherPKD-mediatedfunctions.Inthecardiaccontext, PKDactivityhasbeenimplicatedintheregulationofmyofilament 1 Avkiran,M.,Rowland,A.J.,Cuello,F.andHaworth,R.S.(2008)ProteinkinaseDinthe calcium sensitivity and dynamics [7–9], voltage-gated calcium cardiovascularsystem:emergingrolesinhealthanddisease.Circ.Res.102,157–163 channel activity [46], protection against ischaemia/reperfusion 2 Steinberg,S.F.(2012)RegulationofproteinkinaseD1activity.Mol.Pharmacol.81, injury [47] and insulin resistance [48], and the potential roles 284–291 3 Haworth,R.S.,Goss,M.W.,Rozengurt,E.andAvkiran,M.(2000)Expressionandactivity of FHL proteins in regulating such processes warrant further ofproteinkinaseD/proteinkinaseCμinmyocardium:evidenceforα1-adrenergic investigation. receptor-andproteinkinaseC-mediatedregulation.J.Mol.Cell.Cardiol.32,1013–1023 The possible mechanism(s) through which FHL1 and FHL2 4 Bossuyt,J.,Chang,C.W.,Helmstadter,K.,Kunkel,M.T.,Newton,A.C.,Campbell,K.S., mayregulatePKDactivationalsorequireconsideration.Previous Martin,J.L.,Bossuyt,S.,Robia,S.L.andBers,D.M.(2011)Spatiotemporallydistinct studiesinratcardiacmyocyteshaveshownthatneurohormonal proteinkinaseDactivationinadultcardiomyocytesinresponsetophenylephrineand PKD activation is achieved through the phosphorylation of endothelin.J.Biol.Chem.286,33390–33400 its activation loop, principally by the novel PKC isoform 5 Iwata,M.,Maturana,A.,Hoshijima,M.,Tatematsu,K.,Okajima,T.,Vandenheede,J.R., PKCε [5,21]. Interestingly, proteomic analysis of the cardiac VanLint,J.,Tanizawa,K.andKuroda,S.(2005)PKCε–PKD1signalingcomplexat PKCε signalling complex has identified FHL2 as a component Z-discsplaysapivotalroleinthecardiachypertrophyinducedbyG-proteincoupling receptoragonists.Biochem.Biophys.Res.Commun.327,1105–1113 protein [49]. Moreover, PKCε association with another LIM- 6 Fu,Y.andRubin,C.S.(2011)ProteinkinaseD:couplingextracellularstimulitothe domain protein, enigma homologue, has been proposed to regulationofcellphysiology.EMBORep.12,785–796 play a key role in its substrate targeting [50]. Given that 7 Haworth,R.S.,Cuello,F.,Herron,T.J.,Franzen,G.,Kentish,J.C.,Gautel,M.and ET1- and PE-induced phosphorylation of PKD at Ser744/Ser748 Avkiran,M.(2004)ProteinkinaseDisanovelmediatorofcardiactroponinI was diminished by FHL1 or FHL2 knockdown in the present phosphorylationandregulatesmyofilamentfunction.Circ.Res.95,1091–1099 study (Figure 4), it is plausible that FHL proteins may act as 8 Cuello,F.,Bardswell,S.C.,Haworth,R.S.,Yin,X.,Lutz,S.,Wieland,T.,Mayr,M., scaffolds that facilitate the co-localization of PKCε and PKD Kentish,J.C.andAvkiran,M.(2007)ProteinkinaseDselectivelytargetscardiactroponin IandregulatesmyofilamentCa2+sensitivityinventricularmyocytes.Circ.Res.100, and thus the phosphorylation and activation of the latter, in a 864–873 signal-responsive manner. Additionally, the differential effects 9 Bardswell,S.C.,Cuello,F.,Rowland,A.J.,Sadayappan,S.,Robbins,J.,Gautel,M., of FHL1 and FHL2 knockdown on ET1- compared with PE- Walker,J.W.,Kentish,J.C.andAvkiran,M.(2010)Distinctsarcomericsubstratesare inducedPKDphosphorylationatSer744/Ser748andSer916(Figure4) responsibleforproteinkinaseD-mediatedregulationofcardiacmyofilamentCa2+ suggest that the two FHL proteins might associate with distinct sensitivityandcross-bridgecycling.J.Biol.Chem.285,5674–5682 subcellular pools of PKD downstream of the ET1 and α - 10 Vega,R.B.,Harrison,B.C.,Meadows,E.,Roberts,C.R.,Papst,P.J.,Olson,E.N.and 1 adrenergic receptors. In this context, it is interesting to note McKinsey,T.A.(2004)ProteinkinasesCandDmediateagonist-dependentcardiac hypertrophythroughnuclearexportofhistonedeacetylase5.Mol.Cell.Biol.24, recent evidence which suggests that ET1 and PE may induce 8374–8385 spatiotemporally distinct patterns of PKD activation in cardiac 11 Harrison,B.C.,Roberts,C.R.,Hood,D.B.,Sweeney,M.,Gould,J.M.,Bush,E.W.and myocytes [4]. The potential contribution of FHL isoforms McKinsey,T.A.(2004)TheCRM1nuclearexportreceptorcontrolspathologicalcardiac to such compartmentalized regulation also requires further geneexpression.Mol.Cell.Biol.24,10636–10649 investigation. 12 McKinsey,T.A.,Zhang,C.L.andOlson,E.N.(2000)Activationofthemyocyteenhancer In conclusion, in the present study, we identify FHL1 factor-2transcriptionfactorbycalcium/calmodulin-dependentproteinkinase-stimulated and FHL2 as novel interaction partners for PKD in cardiac bindingof14-3-3tohistonedeacetylase5.Proc.Natl.Acad.Sci.U.S.A.97, myocytes and we show that FHL1 and FHL2 regulate PKD 14400–14405 13 McKinsey,T.A.,Zhang,C.L.,Lu,J.andOlson,E.N.(2000)Signal-dependentnuclear activation in response to neurohormonal stimulation. Although exportofahistonedeacetylaseregulatesmuscledifferentiation.Nature408, such regulation affects HDAC5 phosphorylation, it does not 106–111 control neurohormonal MEF2 activation, suggesting that PKD 14 Lu,J.,McKinsey,T.A.,Nicol,R.L.andOlson,E.N.(2000)Signal-dependentactivation regulationbyFHLproteinsislikelytobeoffunctionalimportance oftheMEF2transcriptionfactorbydissociationfromhistonedeacetylases.Proc.Natl. in PKD-mediated (patho)physiological processes other than Acad.Sci.U.S.A.97,4070–4075 ©(cid:2)c 20T1h4e TAhueth oArusthJoour(rsn)alcompilation(cid:2)c 2014BiochemicalSociety The author(s) has paid for this article to be freely available under the terms of the Creative Commons Attribution Licence (CC-BY) (http://creativecommons.org/licenses/by/3.0/) which permits unrestricted use, distribution and reproduction in any medium, provided the original work is properly cited.

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.