• No results found

Suppression of DNA-dependent protein kinase sensitize cells to radiation without affecting DSB repair

N/A
N/A
Protected

Academic year: 2022

Share "Suppression of DNA-dependent protein kinase sensitize cells to radiation without affecting DSB repair"

Copied!
10
0
0

Loading.... (view fulltext now)

Full text

(1)

ContentslistsavailableatScienceDirect

Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis

jo u r n al ho me p a g e :w w w . e l s e v i e r . c o m / lo c a t e / m o l m u t C o m mu n i ty a dd r e s s :w w w . e l s e v i e r . c o m / l o c a t e / m u t r e s

Suppression of DNA-dependent protein kinase sensitize cells to radiation without affecting DSB repair

Ann-Sofie Gustafsson

, Andris Abramenkovs, Bo Stenerlöw

SectionofBiomedicalRadiationSciences,DepartmentofRadiology,OncologyandRadiationScience,RudbeckLaboratory,UppsalaUniversity, DagHammarskjöldsVäg20,SE-75185Uppsala,Sweden

a r t i c l e i n f o

Articlehistory:

Received9April2014

Receivedinrevisedform4June2014 Accepted16June2014

Availableonline22June2014

Keywords:

DNArepair DNA-PKcs Ionizingradiation DNA-PKdeficiency NU7441

a b s t r a c t

EfficientandcorrectrepairofDNAdouble-strandbreak(DSB)iscriticalforcellsurvival.Defectsinthe DNArepairmayleadtocelldeath,genomicinstabilityanddevelopmentofcancer.Thecatalyticsubunitof DNA-dependentproteinkinase(DNA-PKcs)isanessentialcomponentofthenon-homologousendjoining (NHEJ)whichisthemajorDSBrepairpathwayinmammaliancells.Inthepresentstudy,byusingsiRNA againstDNA-PKcsinfourhumancelllines,weexaminedhowlowlevelsofDNA-PKcsaffectedcellular responsetoionizingradiation.DecreaseofDNA-PKcslevelsby80–95%,inducedbysiRNAtreatment,lead toextremeradiosensitivity,similartothatseenincellscompletelylackingDNA-PKcsandlowlevelsof DNA-PKcspromotedcellaccumulationinG2/Mphaseafterirradiationandblockedprogressionofmitosis.

Surprisingly,lowlevelsofDNA-PKcsdidnotaffecttherepaircapacityandtheremovalof53BP1or␥- H2AXfociandrejoiningofDSBappearednormal.Thiswasinstrongcontrasttocellscompletelylacking DNA-PKcsandcellstreatedwiththeDNA-PKcsinhibitorNU7441,inwhichDSBrepairwereseverely compromised.ThissuggeststhattherearedifferentmechanismsbywhichlossofDNA-PKcsfunctions cansensitizecellstoionizingradiation.Further,fociofphosphorylatedDNA-PKcs(T2609andS2056)co- localizedwithDSBandthiswasindependentoftheamountofDNA-PKcsbutfociofDNA-PKcswasonly seeninsiRNA-treatedcells.OurstudyemphasizesonthecriticalroleofDNA-PKcsformaintainingsurvival afterradiationexposurewhichisuncoupledfromitsessentialfunctioninDSBrepair.Thiscouldhave implicationsforthedevelopmentoftherapeuticstrategiesaimingtoradiosensitizetumorsbyaffecting theDNA-PKcsfunction.

©2014TheAuthors.PublishedbyElsevierB.V.ThisisanopenaccessarticleundertheCCBY-NC-SA license(http://creativecommons.org/licenses/by-nc-sa/3.0/).

1. Introduction

Ionizingradiationandendogenousprocesses,e.g.freeradicals formedinnormal cellularmetabolismor collapseofreplication forks during replication of DNA, may result in complex dam- age to the DNA. Among the different types of DNA damage, DNA double-strand break (DSB) is the most unfavorable type of DNA lesion, and unrepaired or misrepaired DSB can leadto genemutations, chromosomalaberrations,permanentcellcycle arrest, apoptosis, mitotic cell death and cancer development [1–3].

InDSBrepairtwopathwayshavebeenimplicatedtoprocess thebreak,non-homologousendjoining(NHEJ)andhomologous recombination (HR) [4]. Some of the key proteins involved in NHEJareDNA-dependentproteinkinasecatalyticsubunit(DNA- PKcs), Ku70 and Ku80 that together form the DNA-dependent

∗ Correspondingauthor.Tel.:+46184713887;fax:+46184713432.

E-mailaddress:ann-sofie.gustafsson@bms.uu.se(A.-S.Gustafsson).

proteinkinase(DNA-PK)complex[5].DNA-PKcsisamemberofthe phosphatidylinositol3(PI-3)-likekinasefamilythatincludesATM (ataxia-telangiectasiamutated)andATR(ATMandRad3-related) [6]. Besidesfunctioning in NHEJ, DNA-PKcsis alsorequired for V(D)Jrecombinationofimmune-globulingenesandT-cellrecep- torgenes,andtelomerelengthmaintenance[7].NHEJisbelieved toplayamoreimportantrolethenHRinmammaliancells,andis presentatalltimeduringthecellcycle,HRislimitedtolateS,G2 andMphase[8–10].

ProteinsinvolvedintherepairandsignalingofDSBplayavital roleincellsurvivalandhealthyhumanlife[11–13].Manytumors typescharacterizedbygenomicinstabilityarecausedbymutations in DSB-responsivegenes.The expressionofDNA-PKcsin tumor biopsiescanbeboth up-and down-regulatedin specifictumor types[14].SeveralcancertypeswithdecreasedlevelsofbothDNA- PKcsandKu70/80havebeenfound[15–17]whichcouldindicate thatpartialdeficiencyofproteinsinvolvedinDSBrepairmaycon- tributetoincreasedriskofdevelopingcancer[14,18].Incontrast, elevatedlevelsofDNA-PKcshavebeenfoundinvarioustumortypes andlinkedtopoorsurvival[19].

http://dx.doi.org/10.1016/j.mrfmmm.2014.06.004

0027-5107/©2014TheAuthors.PublishedbyElsevierB.V.ThisisanopenaccessarticleundertheCCBY-NC-SAlicense(http://creativecommons.org/licenses/by-nc-sa/3.0/).

(2)

2 A.-S.Gustafssonetal./MutationResearch769(2014)1–10

Besidesitsdirectroleinrepair,itisbecomingevidentthatDNA- PKcsmayhaveotherfunctionsthatpromotecellsurvival.Arlander etal.(2008)suggestedthatDNA-PKcscontributestotheG2check- pointsandcellcycleregulation[20].Recentlyitwasalsoshown thatDNA-PKcsisinvolvedinstabilizationofthemitoticspindle formationinawaythatreducedlevelsofDNA-PKcsmaydirect thecelltowardmitoticcatastrophe[21–23].Furtherdataindicates thatDNA-PKcsisacentralplayerinG2progressionandlowlev- elsofDNA-PKcspromotesafastprogressionfromSphasetoG2, butinterfereswithprogressionthroughmitosisandleadstocell accumulationinG2[22].HumancellsdeficientinDNA-PKshowa severesensitivitytoradiationandreducedcapacitytorepairDSBs [11,24,25]anditispossiblethatthisinabilitytoprogressthrough thecell-cycleisaffectedbythepresenceofunrepairedDNA[26].

Inthepresentinvestigation,westudiedDNArepairincellswith reducedlevelsofDNA-PKcs.HumancellstransfectedwithsiRNA targetingDNA-PKcs,resultingin5–20%oftheoriginalDNA-PKcs content,exhibitedsimilarextremeradiosensitivityascellscom- pletelylackingDNA-PKcs.Surprisingly,thelowDNA-PKcs-levels hadnosignificanteffect onDSBrepair capacity, butlow levels of DNA-PKcs affected thecell cycle, resulting in siRNA treated cellaccumulationinG2afterirradiationandinabilitytoproceed throughmitosis.Thesedatasupportrecentfindingssuggestingthat DNA-PKcsisanimportantregulatorofthecell-cyclewhilewehere showthatthisfunctionseemstobeuncoupledfromthecellular capacitytorepairDSBs.

2. Materialsandmethods 2.1. Celllinesandcultureconditions

HumanepithelialcancercelllineA431(ATCC,Manassas,VA, USA) was grown in Ham’s F-10. Human oral squamous carci- nomacell line H314 (ATCC) was grown in DMEM/Ham’s F-12.

HumancolorectalcarcinomacelllineHCT116(ATCC)wasgrownin McCoy’s.NormalhumanskinfibroblastGM5758cellline(Human GeneticMutantCellRepository,Camden,NJ,USA)wasgrownin MEM, supplemented with 2× concentration of MEM vitamins, essentialandnon-essentialamino acids.Thehumangliomacell lines M059J deficient in DNA-PKcs (ATCC) and M059K (ATCC) withwild-typeDNA-PKcswereculturedinDMEM/Ham’sF12sup- plementedwithnon-essential amino acids.Culturemedium for allthecells was supplemented with10%FCS (Sigma, St. Louis, MO),2mMl-glutamine,100IU/mlpenicillinand10␮g/mlstrep- tomycin(BiochromKg,BerlinGermany).Allcellswereculturedin ahumidifiedincubatorwith5%CO2at37Candtrypsinizedwith trypsin–EDTA(0.25%trypsin,0.02%EDTA,BiochromKg).

2.2. Irradiationandinhibitortreatment

Cellswereirradiatedwith137Cs␥-rayphotons(Gammacell40 Exactor,MDSNordion,Kanata,Canada)atadoserateof∼1Gy/min.

NU7441wasobtainedfromSelleckchem(Houston,TX).NU7441 wasdissolvedindimethylsulfoxide.Fortreatment,cellswerepre- treatedin37Cfor1hwith2–10␮MNU7441beforeirradiation.

ClonogenicsurvivalassayNU7441wasremoved24hafterirradi- ationwithfreshmedium.Dimethylsulfoxidewasusedin mock treatedcells.

2.3. Antibodies

Primarymouse-monoclonalantibodiesusedwere,DNA-PKcs (ab1832,Abcam), p-DNA-PKcs (Thr2609)(ab18356, Abcam), ␤- actin(Sigma)and ␥-H2AX(Millipore).Primaryrabbit-polyclonal antibodyusedwere53BP1(BethylLab,Montgomery,TX),p-DNA- PKcs(Ser2056)(ab18192,Abcam),p-DNA-PKcs(Thr2609)(Santa

Cruz),p-AKT(Ser437)(CellSignaling)andp-HistoneH3(Millipore).

NucleuswasstainedwithDAPI(Invitrogen).Secondaryantibod- iesAlexaFluor488goatanti-mouseandAlexaFluor555donkey anti-rabbit(Invitrogen).

2.4. siRNAtransfection

ForthesiRNAtransfectionofDNA-PKcsaSMARTpoolprovided byDharmacon(ThermoScientific)wasusedwithfoursiRNAspe- cificforPRKDC(catalogueno.L-005030-00-0020)andaSMARTpool asnon-specific control(mock) (catalogueno.D-001810-10-20).

TransfectionswereperformedwithDharmaFECT1 Transfection Reagent(cataloguenoT-2001-01)according totheinstructions ofthesupplier.TheconcentrationofsiRNAwas100nMandwas addedtothecellstogetherwithDharmaFECTI.Cellswereseeded onedaypriortransfection,andcellswereanalyzedthreedaysafter transfection.

2.5. SDS-PAGEandWesternblotanalysis

Cellswerelysedinicecoldlysisbuffercontaining1%Tween-20, 20mMTris(pH8.0)137mMNaCl,10%glycerol,2nMEDTA,1mM activatedsodiumorthovanadate(Sigma) and protease inhibitor cocktail (P8340, Sigma). Cell extracts were loaded and elec- trophoresedonNuPAGENovex3–8%Tris–acetategels(Invitrogen).

Wettransfertonitrocellulosemembraneswasdoneovernightand thenthemembraneswereblockedfor1hin5%BSA,PBSandthen incubatedwiththeprimaryantibodyovernightat4C.Secondary antibodies(626520and656120)(Invitrogen)labeledwithHorse RadishPeroxidase wereincubatedfor1hatroomtemperature.

ImmunoreactivebandswerevisualizedinaCCDcamera(SuperCCD HR,Fujifilm,Japan)aftertreatmentwithelectrochemiluminescent solution(Immobilon). The intensity ofthe bands wasanalyzed usingImageJsoftware(NIH,US).

2.6. Cellsurvival

Cellswerecountedandanappropriatenumberwasseededinto T-25cellflaskintriplicateforeachdose.Cellswereirradiatedwith 1–4Gy,4hafterseeding.After1–2weekscellswerefixedin95%

EtOHandstainedwithMayer’shematoxylinandcolonieswith>50 cellswerescored.

2.7. Mitoticanalysis

ThreedaysaftertransfectionwithsiRNAcellswereirradiated with5Gy,andlettorepairfor24hinfreshmediumbeforefixation andimmunostainingwithp-HistoneH3 (p-H3)antibody.Atthe microscopep-H3positivecellswerescored,atleast>250cellsper datapoint.

Fornocodazoletreatment,cellsweretreatedwith1␮l/mgnoco- dazole(Sigma),3daysaftertransfectionwithsiRNA,andincubated for6h.After6h,nocodazolewasremovedandcellswereirradiated with2Gyandlettorepairfor18hinfreshmediumbeforefixation andimmunostainingwithp-H3antibody.Atthemicroscopep-H3 positivecellswerescored,atleast>400cellsperdatapoint.

2.8. Immunofluorescencestainingandquantificationoffoci

Cellsweregrownonmicroscopeslidesforthreedaysaftertrans- fectionandwereirradiatedwith1–5Gy.AfterrepairofDNAfor varioustimesat37C,thecellswerefixedinmethanolfor20min at−20Cfollowedbypermeabilizationinicecoldacetonefor10s.

Blockingwasperformedin10%FCS-PBSfor1hatroomtemperature andthenincubationovernightat4Cwithprimaryantibodies.Sec- ondaryantibodieswereincubatedfor1hat37C(dilution1:400).

(3)

Fig.1. ReductionofDNA-PKcsleadstoextremeradiosensitivity.(A)WB:expressionofDNA-PKcsaftersiDNA-PKcstransfectionfor3daysinA431,HCT116,H314,GM5758 cellsandinDNA-PKcsdeficientM059Jcells.QuantificationofbandintensitywithImageJ,fractionofresidualDNA-PKcsisshownforeachcellline,±SDfromthreeexperiments.

(B)RepresentativeimagesofA431cellmock-transfectedandsiDNA-PKcstransfectedfor3days,fixedandimmunostainedwithantibodiesagainstDNA-PKcs.Nucleiwere visualizedwithDAPIstaining.(C)Clongenicsurvivalinthreecelllines,A431,HCT116andH314,transfectedwithsiRNAagainstDNA-PKcs(siDNA-PKcs)for3daysortreated withnon-specificsiRNA(mock).Incomparison,survivalforM059Jcells(lackingDNA-PKcs)andM059K(wtDNA-PKcs)areshown.Colonieswith>50cellswerescored10–15 daysafterirradiation.SF=survivingfraction.Datarepresentmean±SDofatleastthreeindependentexperimentsforeachcellline.(D)Radiationsensitivity,summaryof meansurvivingfractionat2Gy.

Slides werecapturedwithZeiss LSM510Meta confocalmicro- scope.ImageswereprocessedandfociwerecountedusingImage Jsoftware(NIH,US).

2.9. DSBRejoiningandpulsed-fieldgelelectrophoresis

Cells were labeled with 2kBq/ml [methyl-14C] thymidine (PerkinElmer),togetherwithsiRNAtransfection,threedaysprior toirradiation.Cellswereirradiatedwith40Gyandembeddedin 90␮lagaroseplugs(0.6%w/v,InCert,Cambrex).Cellplugswere lysedusingatwo-stepcoldlysisprotocolinESPlysisbuffer(2%N- lauroylsarcosine(Sigma),1mg/mlProteinaseK(Roche),alldiluted in0.5MEDTA(Na3)atpH8.0[27].After>20hthecellplugswere movedtoHS-bufferandincubatedovernightat4C(HS:HighSalt;

1.85MNaCl,0.15MKCl,5mMMgCl2,2mMEDTA,4mMTris,0.5%

TritonX-100,pH7.5,TritonX-100isaddedjustbeforeuse).Plugs werewashedtwicein0.1MEDTAandoncein0.5×TBEat4C prior toelectrophoresis.Theplugswerethen loadedintowells inachilled(4C)agarosegel(0.8%SeaKemGold,Lonza).Thegel wasplacedintoaPFGEunit(GeneNavigator,AmershamPharmacia Biotech,Uppsala,Sweden)with120betweenthefields.Following electrophoresisin0.5×TBEat10C,thegelswereslicedattheposi- tionofthe5.7MbpchromosomefromSchizosaccharomycespombe (BioRad),and14Cinthegelsegmentsweremeasuredbyliquidscin- tillation.ThefractionofradioactivitycorrespondingtoDNAofsize lessthan5.7Mbpwasdividedbythetotalradioactivityinthelane, givingtheFractionActivityReleased(FAR)whichisameasureof

DNAdouble-strandbreaks.Datawasnormalized,setto100%at t=0.

3. Results

3.1. ReductionofDNA-PKcsleadstoextremeradiosensitivity

ThereductionofDNA-PKcswasconfirmedonWesternblotper- formedinparallelwitheachexperiment(Fig.1A).Proteinlevels weremarkedlydecreasedtwodaysfollowinginitialtransfection andwerestableforatleastfourdays(Supplementary Fig.S1A).

Thetransfection efficiencyvariedslightly betweenexperiments andcelllines,buttherewasalarge80–95%reductioninDNA-PKcs levelsinallofthetestedhumancelllines,A431epithelialcancer cells,H314oralsquamouscarcinomacells,HCT116colorectalcar- cinomacellsandGM5758normalfibroblasts.Thedepletionwas alsoevidentonimageswithimmunofluorescentstainingagainst DNA-PKcs(Fig.1B).ATMlevelswerealsoanalyzedforpotential lossasaconsequenceofDNA-PKcsknock-down,howeverATMlev- elswerenotaltered(SupplementaryFig.S1B).TheM059Jglioma celllinewhichcompletelylacksDNA-PKcsissensitivetoionizing radiationandisshownforcomparison.Totestwhetheran80–95%

depletionofDNA-PKcsinfluencethesurvivalandproliferationof cells,aclonogenicsurvivalassaywasperformedonthreediffer- entcelllines,A431,H314andHCT116(Fig.1C).siRNAmediated depletionofDNA-PKcsmarkedlyreducedthesurvivingfractionin allcelllinesandatadoseof2Gytherewasa3–10foldreduction

(4)

4 A.-S.Gustafssonetal./MutationResearch769(2014)1–10

Fig.2. CellswithlowlevelsofDNA-PKcsaredelayedinthemitoticprogression.(A)AccumulationofmitoticcellsinasynchronousA431cellswithlowlevelsofDNA-PKcs.

Cellswereirradiatedwith5Gyandscoredforp-H3-positivecells24hafterirradiation.Datarepresentmean±SDoftwoindependentexperiments.Atleast250cells/time pointwerescoredperexperiment.(B)SuppressionofDNA-PKcsleadstoaccumulationofmitoticcellsfollowingirradiationandtreatmentwithnocodazole.Cellswere synchronizedfor6hwithnocodazoleandirradiated,andafter18hincubationinmediumwithoutnocodazole,cellswerefixedandp-H3-positivecellswerescored.Ctr cellswereunirradiated.Datarepresentmean±SDoftwoindependentexperiments.Atleast400cells/timepointwerescoredperexperiment.(C)PhosphorylatedDNA-PKcs (Thr2609)isdislocatedfromchromatininmitoticcellsandlocatedatthemidbody(whitearrows).Thr2609(green)ismergedwithp-H3-positive(red).Incomparison,total DNA-PKcs(green)ininterphaseandmitoticcellsareshown(right).NucleiwerestainedwithDAPI(blue).

inthesurvival(Fig.1CandD).Incomparison,M059Jcells,which completelylacksDNA-PKcs,areshown.Thus,partialreductionin DNA-PKcsmayleadtosimilarextremeradiosensitivityasseenin cellscompletelylackingDNA-PKcs.

3.2. CellswithlowlevelsofDNA-PKcsaredelayedinthemitotic progression

Recentdata suggest that DNA-PKcs, beyonditsrole in DNA repair,mayhaveothercellularfunctionsandcouldbeanimpor- tantregulatorofthecell-cycle[21–23].Totestthehypothesisthat lowDNA-PKcslevelscouldleadtoaberrantmitosis,asynchronous A431cellswerescoredforphosphorylatedHistoneH3(p-H3)24h afterirradiation.p-H3isrelatedtoseveralphasesofmitosisand

usedasamarkerhereinforpositiveG2/Mcells.Cellstreatedwith siRNAagainstDNA-PKcsexhibitedtwo-foldincreaseofp-H3posi- tivecellswhencomparedtomocktreatedcells(Fig.2A).Further,in siRNAtreatedcellsasubstantialnumberofthemitoticcellswere inlatemitosis(metaphase,anaphaseortelophase),whichwasin strongcontrasttomocktreatedcells(datanotshown).Toenrich thefractionofcellsinmitosis,cellsweretreatedwithnocodazole, anagentusedtoarrestandsynchronizecellsinG2/M-phase.Cells wereincubatedfor6hwithnocodazole,irradiatedandreleased for18hbyremovingthedrugbeforefixationandscoringofp-H3- positivecells.Afterirradiationwith2Gytherewasa2-foldincrease in the fractionof p-H3-positive cells when treated with siRNA againstDNA-PKcscomparedtomocktreatedcells(Fig.2B).Thus, theseresultssuggestthatlowamountsofDNA-PKcscouldleadto

(5)

accumulationofcellsinG2/Mphaseafterirradiation.Theresults indicatethatirradiatedcellswithlow levelsof DNA-PKcsenter mitosisascellswithnormalproteinlevels,butarethendelayed inthemitoticprogression.Thisfurtherstrengthensthehypothesis that DNA-PKcs hasa role in regulation of the mitotic progres- sion.Indeed,inthemitoticA431cells DNA-PKcswasdislocated fromchromatinandmitosis-specificDNA-PKcs phosphorylation (Thr2609)appearedtoconcentrateattwositesonoppositeside ofthenucleusimplyingadifferentrolethaninDSBrepair(Fig.2C), although there was no evident difference in the protein dis- tribution between siDNA-PKcs or mock treated cells (data not shown).

3.3. CellswithlowresiduallevelsofDNA-PKcshaveapparently normalDSBrepair

SincecellswithdefectiveDNA-PKhaveareducedrepaircapac- ity,itisreasonabletoassumethattheextremeradiosensitivityand themitoticfailure,seeninDNA-PKcsdepletedcellsabove,canbe explainedbyinabilitytorepairDSB.Totestthishypothesis,the formationanddisappearanceof53BP1(p53bindingprotein1)foci and␥-H2AXwasanalyzedinsinglecellsafterirradiation.GM5758 andA431cellsweretransfectedwithsiRNAagainstDNA-PKcsor mocktreatedandafter3dayscellswereirradiatedandincubated forrepair.53BP1fociwererapidlyformedinbothcelllinesafter irradiationand theinitial numberof 53BP1 fociwas similarin cellstransfectedwithsiRNAagainstDNA-PKcsandmocktreated (Fig.3A).However,somewhatsurprising,thereducedamountof DNA-PKcsdidnotsignificantlyaffectthekineticpropertiesofeither 53BP1or␥-H2AXineithercellline(Fig.3BandD)and1–24hafter irradiationthereweresimilarnumbersoffociincellstransfected withsiRNAagainstDNA-PKcsand mocktreatedcells. Similarly, therewasnodifferencebetweensiDNA-PKcs andmock treated cellswhentheresidualnumberof53BP1fociwasscored24hafter irradiationwithdifferentdoses(Fig.3C).Theseresultsshowthat siDNA-PKcsdoesnotaffecttheformation anddisappearanceof boththeDSBmarker53BP1and␥-H2AXafterirradiation.

TofurtherinvestigateiflowDNA-PKcslevelsaffectthecapacity torepairradiation-inducedDSBs,DSBrejoiningwasanalyzedusing pulsed-fieldgelelectrophoresis(PFGE).Despitethelargereduction inthelevelsofDNA-PKcs,therewasnoeffectonDSBrejoiningin anyofthefourtestedcelllines(Fig.3E),notevenforradiationdoses upto200Gy,whichinducesatleast5000–6000DSBpercell(Sup- plementaryFig.S2). Incontrast,M059Jcells,completelylacking DNA-PKcs,showedonlymarginalrejoiningupto24hpostirra- diation(Fig.3E).Insummary,thesedatasuggestthatdespitelow residuallevelsofDNA-PKcs,i.e.5–20%oftheoriginalproteinlevels, cellshaveapparentlynormalDSBrepair.

3.4. InhibitionoftheDNA-PKcskinaseactivitysensitizecellsto radiationandreduceDSBrepair

BecausesiDNA-PKcstreatmentmarkedlyreducedcellsurvival afterirradiation,despiteanefficientDNArepair,wewantedtocom- paresiDNA-PKcstreatedcellswithcellstreatedwiththespecific DNA-PKcsinhibitorNU7441.Cellsurvivalwasmarkedlyreduced inA431cellstreatedwithNU7441(Fig.4A),andthedose-response wassimilartothatinM059Jcells,lackingDNA-PKcs(Fig.1C).We nextinvestigatedwhethertreatmentwithNU7441affectedthefor- mationanddisappearance53BP1inA431andGM5758cells.The initialnumberof53BP1fociafter1hwassimilarinuntreatedcells andcells treatedwithNU7441,however,after4–24hofrepair, theresidualnumbersof53BP1fociwerehigherin theNU7441 treatedcells(Fig.4B).Further,inGM5758cellsDSBrejoiningwas almostcompletelyinhibitedbyNU7441(Fig.4C).EvidentlyDSB repairisdifferentiallyaffectedwhethertheactivityofDNA-PKcs

is inhibited, or ifthere are low amountsof DNA-PKcs present.

OurdatashowthatinhibitionofDNA-PKcsactivityreduceboth DSBrepairandcellsurvival afterexposuretoionizingradiation (Fig.4).Incontrast,loweringofDNA-PKcslevelsviasiRNAtreat- mentmarkedlyreducedcellsurvivalwithoutanycleareffectson repair(Figs.1and3).Thus,thepresentdataimplicatethatthereare differentmechanismsbywhichlossofDNA-PKcsfunctionsensitize cellstoionizingradiationandsuggestthatDNA-PKcshasacritical roleinmaintenanceofsurvivalafterradiationexposure,besidesits importantfunctioninDSBrepair.

AnumberofstudiessuggestacloseinteractionbetweenAKTand DNA-PKcs[28,29].TreatmentwithNU7441orsiDNA-PKcsabol- ishedtheactivityofAKT(Ser473)afterirradiation(Supplementary Fig.S3A),supportingtheroleofDNA-PKcsinactivationofAKTand thatlowamountsofDNA-PKcs,viadownregulationofAKT,may promoteapoptosis.Totestthis,cellswereirradiatedandscoredfor apoptosisbymeasuringcleavedPARP.Comparedtounirradiated cellstherewasaclearincreaseinPARPcleavage72hafterirradi- ationwith8Gy,buttherewasnodifferencebetweensiDNA-PKcs andmocktreatedcells(SupplementaryFig.S3B).

3.5. PhosphorylationofDNA-PKcsatThr2609andSer2056

ATMandDNA-PKcsarethemajorkinasesactivatedfollowing radiation.DNA-PKcsisrapidlyactivatedbybothphosphorylation andautophosphorylationuponirradiationandseveralphosphor- ylation sites have been identified so far. ATM is essential for phosphorylation of Thr2609 [30] and DNA-PKcs is responsible forSer2056phosphorylation[31].Westernblotanalysiswasper- formedonA431cellstotestifradiation-inducedphosphorylation ofThr2609andSer2056areaffectedbyadepletionofDNA-PKcs.

The overall phosphorylationof both Thr2609 and Ser2056 was almostcompletelyabolishedincellstargetedwithsiRNAagainst DNA-PKcs(Fig.5A),mostlikelyduetodecreasedlevelsofDNA- PKcs.However,cellsinhibitedwiththeDNA-PKcsinhibitorNU7441 showednodifferenceinDNA-PKcsphosphorylation.

Despite the reduced amount of phosphorylated DNA-PKcs in Western blot,formation and disappearance of Thr2609 and Ser2056fociafterirradiationweresimilarinmock-andsiRNA- treated cells, and thesefoci co-localizedwiththe DSBmarkers 53BP1and␥-H2AX,indicatingthatthephosphorylationofDNA- PKcsatDSBsiteswerenotaffectedbytheamountofDNA-PKcs (Fig.5BandC).Further,noapparentdifferencecouldbeseeninfoci intensitybetweensiDNA-PKcsandmocktreatedcells.Theseresults suggestthattheproteinsleftinthecellsaregatheringaroundDSB andarefullyactivatedatthesesites.CellstreatedwiththeDNA- PKcsinhibitorNU7441exhibitedanapparentlynormalformation ofThr2609andSer2056fociatearlytime-pointsafterirradiation.

However, 4–24h post-irradiation,thenumbers of Thr2609 and Ser2056focishowedonlyasmalldecrease(Fig.5C),whichcoin- cidedwiththe53BP1analysisandisinlinewiththehypothesisthat ATMactivationpromotesDNA-PKcsbindingtoDNAbutinhibition byNU7441preventsDNA-PKcstoparticipateinDSBrepair.

Interestingly,fociofDNA-PKcswereclearlyvisible60minafter irradiationofcellstreatedwithtargetsiRNA(Fig.6).TheDNA-PKcs focioverlappedwiththeDSBmarker53BP1andthesefociwere notseenincellswithnormallevelsofDNA-PKcs(mock-treated).

ThiscouldbeexplainedbythehighlevelsofDNA-PKcsinnormally presentinhumancellsbutwhentheproteinlevelisdecreasedby 80–95%theallocationaroundDSBofactiveproteinscanbeseen.

Notably,theDNA-PKcsfocibecamevisible60minafterirradiation andafter4–24htheybecamemoredistinct,whereascellsanalyzed atshorterrepairtimesjustdisplayedaweakersignalofDNA-PKcs overthewholenucleus.Thus,lowlevelsofDNA-PKcsmaycausea delayintheredistributiontotheDSBsites.

(6)

6 A.-S.Gustafssonetal./MutationResearch769(2014)1–10

Fig.3.CellswithlowresiduallevelsofDNA-PKcshaveapparentlynormalDSBrepair.(A)representativeimagesofA431andGM5758cellsmocktreatedorsiDNA-PKcs transfected,irradiatedwith2Gyor1Gy,respectively,andallowedtorecoverfortheindicatedtimes.0Gywasusedascontrol.Cellswerefixedandimmunostainedwith 53BP1(red)andnucleiwerestainedwithDAPI(blue).(B)Kineticsof53BP1fociincellstreatedasin(A).Atleast100cells/timepointswerescoredforfoci.Datarepresent mean±SDofthreeindependentexperiments.(C)Doseresponsebyaveragenumberof53BP1focipercell.CellswereeithermocktransfectedorsiDNA-PKcstransfectedand thenirradiatedatindicateddosesandallowedtorecoverfor24hbeforefixationandimmunostaining.Countingandstatisticsasin(B).(D)Timeresponseof␥-H2AXfociin cellstreatedasin(A).Countingandstatisticsasin(B).(E)RejoiningofDSBsinA431,HCT116andH314cellsmock-transfectedorsiDNA-PKcstransfectedbeforeirradiation.

CellswereallowedtorecoverfortheindicatedtimespriortoPFGEanalysis.M059KandM059Jdataareshownascomparison.DatawerenormalizedtotheDSBlevelat t=0handeachdatapointrepresentsmean±SDofthreeindependentexperiments.

4. Discussion

ItiswellknownthatdecreasedcapacitytorepairDNAdamage byradiationorotherDNAdamagingagentsisacancerriskfactor andDNA-PKhasbeenimpliedtobedownregulatedinmanydiffer- enttumortypes.Theexpressionseemstobetissuedependentand ithasbeendemonstratedthattissuefromuterine,breastandlung

cancerhaveatendencyoflowDNA-PKlevelswhereasheadand neckcancershownormallevels[15,16,32].Humancellsmaintain ahighreservoirofproteinsinvolvedinNHEJofDSBrepairandhigh levelofproteinsallowsthecelltoprotectthegenomebytheimme- diatepresenceofrepairproteinsatthebreaks,minimizingthetime forassemblyandactivationatthebreaksite.Therearenearlyhalfa millionKumoleculesandaroundonehundredthousandDNA-PKcs

(7)

Fig.4.InhibitionoftheDNA-PKcskinaseactivitysensitizecellstoradiationandreduceDSBrepair.(A)ClonogenicsurvivalinA431cellstreatedwiththeDNA-PKcsinhibitor NU7441(2␮M).CellswerepreincubatedwithNU74411hbeforeirradiationatindicateddoses.NU7441wasremoved24hafterirradiationandcolonieswith>50cellswere scoredafter10days.(B)53BP1fociformationandremovalinA431andGM5758cells.Cellswereincubatedwith10␮MNU74411hbeforeirradiationatindicateddosesand allowedtorecoveratindicatedtimesbeforefixationandimmunostaining.Fortimeresponsecellswereirradiatedwith2Gy,andfordoseresponsefociwerecountedafter 24h.Atleast100cells/timepointswerescoredforfocianddatarepresentmean±SDofthreeindependentexperiments.(C)RejoiningofDSBsinGM5758cellstreatedwith 5␮MNU7441.CellswereincubatedwithNU74411hpriortoirradiationwith40GyandallowedtorecoverfortheindicatedtimesbeforepreparationandanalysisbyPFGE.

Datawasnormalized(t=0h)andeachdatapointrepresentsmean±SDofthreeindependentexperiments.

moleculespercell[33].Thedatapresentedhereprovidefurther indicationsoftheimportanceofDNA-PKcsforpromotingcellular survival.Irrespectiveofthestrongradiosensitizingeffect,weshow thatwithlevelsofDNA-PKcsbelow20%comparedtonormalcells, cellsretaintheirabilitytorejoinDNAfragmentsandremove53BP1 and␥-H2AXfociwithnormalspeed.Toourknowledge,thepresent studyisthefirsttoshowthatknock-downofcriticalDSBrepairpro- teinsleadstoextremeradiosensitivitywithoutanyapparenteffect ontherepair.Thus,thehighlevelsofDNA-PKcsinnormalcells mayindicateimportantrolesinothercellularfunctionsandthatan excessofmoleculesneedtobepresentinthestressresponseupon severeDNAdamage.Ourobservationsshowthatafterirradiation, cellswithlimitedlevelsofDNA-PKcshaveatwo-foldincreasein thefractionofcellsarrestinginG2/M,indicatingthatlowlevelsof DNA-PKcsmaystallmitosis.Thiswasbothshowninsynchronized andasynchronouscells(Fig.2).Theseobservationsareinlinewith astudybyShangetal.,showingthatDNA-PKcscontributestothe normalspindleformationandcentrosomestabilityaswellasinac- tivationofDNA-PKcscouldcausemultipolarspindleandmitotic catastropheafterDNAdamage[21].WeherealsoshowthatDNA- PKcsisdislocatedfromthechromatininthemitoticnucleusand aDSB-independentThr2609phosphorylationwasconcentratedat twodistinctsitesoneachsideofthechromatin(Fig.2C),indicat- ingaroleofDNA-PKatcentrosomesinthemitoticcell.Thisisin linewithrecent datashowingco-localizationofphosphorylated DNA-PKcs(Thr2609)andPlk1inmitoticcells[21–23,34].Deple- tionofthismitoticphosphorylationofDNA-PKcscouldresultin delayanddysfunctioninthemitotictransition.Thiscouldleadto alossorgain ofawholechromosome,which isthemostcom- monchromosomalinstabilityassociatedwithhumancancers[23].

ItwouldberationaltoassumethatthepresenceofunrepairedDSB inDNA-PKcsdeficient/suppressedcellsshouldcausedelayincell- cycleprogressionandmitoticfailure.Importantly,ourdatasuggest that DNADSB repair capacity isnot thelimiting factor in cells withlowDNA-PKcslevels.Thisinformationcouldbecrucialforthe understandingofthecomplexityofDNAdamagestressresponse, and specifically,themultifunctional roles ofDNA-PK. ATM and ATRhavebeenfirmlyestablishedascentralplayersintheactiva- tionofthecheckpointpathwaysthatrelayontheDNAdamage

signaling to downstream effectors through phosphorylation [35,36] and perhaps DNA-PK plays a part in this role as well.

InhibitionofPI-3kinasesincreasemitoticarrestandmitoticcell death[37]and ATM,ATRandDNA-PK haveall beenconfirmed tolocalizetocentrosomesduringmitosis[34].Thissuggeststhat thesePI3-likekinasescomplementeachotheroncentrosomesand thefollowingdownstreamphosphorylationsarenecessaryforcell cycleprogressionandsynthesisofmicrotubules[34].Wecannot excludethepossibilityofalternativepathways(e.g.homologous recombination)mayrejointheDSBs,howeverthisdoesnotexplain thearrestofcellsinG2/Mphase.

CellswithtotaldeficiencyofDNA-PKcs,liketheM059Jcellline, displayahypersensitivitytoionizingradiationthatreflectsacrucial roleofthisenzymeinmaintainingrepaircapacity[24,25,38,39]and chromosomestability[40].PhosphorylationofDNA-PKcsiscritical forthebindingandreleasefromtheDNAend andinhibitionof DNA-PKcswillresultinblockedDNAends,whichinhibitfurther processingandefficientligation.Themajorityofphosphorylation sitesarelocatedattheABCDE-andPQRclusterandmutationsat eitherofthesesitesimpairsDNArepair[30,35,39,41–44].There- fore,targetingandinhibitingDNA-PKactivityisanattractivecancer therapystrategyandthemostsuccessfulapproachhasbeenwith theuseofsmallmoleculestargetingtheATPbindingsiteofthe kinasedomain[45].A promisingdrugis theNU7441,based on theLY294002backbone,whichshowsastronginhibitionofDNA- PKcsandDNArepairincells[46].InhibitionbyNU7441doesnot resultinthesameradiosensitivityascellstotallylackingDNA-PKcs [47],howeverweobservedanalmostcompleteinhibitionofDNA repairincellstreatedwithNU7441,whichwasinstrongcontrastto cellswithdecreasedlevelsofDNA-PKcs.Eventhoughtheinhibitors holdpromisingresultsforimprovingcancertherapy,themolecules knownsofararelimitedbytheirpoorpharmacokinetics[45,48].

Further,ourdataimplicatesthatsuppressionofDNA-PKcscouldbe apreferabletherapeuticstrategy;whereasinhibitionofthekinase activitybyNU7441blockstheDSBrepairinbothdividingandnon- dividingcells (Fig.4), whilesuppressionof DNA-PKcsbysiRNA mainlyaffectsthedividingcells.

ThehighsensitivitytoionizingradiationincellswithlowDNA- PKcslevelscouldbeduetodysregulationofdownstreammolecules.

(8)

8 A.-S.Gustafssonetal./MutationResearch769(2014)1–10

Fig.5.PhosphorylationofDNA-PKcsatThr2609andSer2056.(A)WB:relativelevelsofDNA-PKcs,p-Thr2609(DNA-PKcs),p-Ser2056(DNA-PKcs)inA431cells,mock- transfected,siDNA-PKcstransfectedortreatedwith10␮MNU74411hbeforeirradiationwith2Gy.Cellswereallowedtorepairfor1hbeforepreparationforWB.Equal loadingwasconfirmedby␤-actinimmunoblot.(B)RepresentativeimagesofA431cells,mock-transfected,siDNA-PKcstransfectedortreatedwith10␮MNU74411hprior toirradiationwith2Gy.Cellswereallowedtorepairfor1hor24hbeforefixationandimmunostainingwitheitherp-Thr2609(DNA-PKcs)(green)andco-stainedwiththe DSBmarker53BP1(red),orp-Ser2056(DNA-PKcs)(red)andco-stainedwiththeDSBmarker␥-H2AX(green).Co-localizationbetweenthedifferentantibodiesisdepicted inthemergedimagesasyellow,nucleiwerestainedwithDAPI(blue).(C)Averagenumberofp-Thr2609(DNA-PKcs)andp-Ser2056(DNA-PKcs)fociinA431andGM5758 cellsmock-transfected,siDNA-PKcstransfectedortreatedwith10␮MNU7441followedbyirradiationatindicateddosesandallowedtorecovertheindicatedtimes.Data representmean±SDofthreeindependentexperiments.Atleast100cells/timepointwerescoredperdatapoint.

Indeed,AKTphosphorylation(Ser473)decreasedinsiRNAtreated cellsafterirradiation.However,increasedapoptosiswasobserved in both, DNA-PKcs silenced cells and mock treated cells when cleavedPARPpathwaywasstudied.(SupplementaryFig.S3).Fur- ther,wecouldnotdetectanysignsofacceleratedsenescence(data notshown), which wasrecently reportedwhen DNA-PKcswas blockedwiththePI3-inhibitorNVP-BEZ235incombinationwith radiation[49].

InourstudyweshowthatincellswithlowlevelsofDNA-PKcs therewasclearvisualizationofDNA-PKcsfocithatoverlappedwith 53BP1,whichwasnot seeninmocktreatedcells. However,the formationofDNA-PKcsfociwasrelativelyslowandthiscouldindi- catea delayinthere-localizationof DNA-PKcstotheDSBsites

insiRNA-treatedcells. Wecouldnotidentifyadifferenceinthe numbersofThr2609andSer2056focibetweencellswithlowDNA- PKcslevelsandcellswithnormalDNA-PKcscontent,howeverin NU7441treatedcellsthemajorityoffociretainedevenafter24h repair.Thisimpliesthattheproteinsleftareactiveandrepairing DSBasinnormalcells,butinhibitionofDNA-PKcskinaseactivity leavesthecellunabletorejoinDNAends.Ourobservationsalso showare-localizationofp-DNA-PKcsduringmitosis,indicatinga phosphorylationeventthatmaybecriticalformitosis.

Insummary,ourdatashowthatDNA-PKcsisnecessaryforreg- ulationof mitoticprogressionafterirradiation,and thisprocess seemstobeindependentofitsroleinDSBrepair.Thisincreaseof radiationsensitivity,comparedtonon-dividingcells,couldbean

(9)

Fig.6.LowlevelsofDNA-PKcsdisplayfociformation.Representativeimagesof A431cellsmock-transfectedorsiDNA-PKcstransfected.Cellswereirradiatedwith 2Gy,fixedandimmunostainedafter15minor4hwithDNA-PKcs(green)andthe DSBmarker53BP1(red).Thetwoimagesweremergedandco-localizationofDNA- PKcsand53BP1aredepictedasyellow,nucleiwerestainedwithDAPI(blue).

advantageinradiotherapyoftumors.Identificationofadditional functionsofDNA-PKcs,beyonditsstrongroleinDSBrepair,could giveimportantknowledgeaboutregulationofgenomicstability andthiscomplexitywouldbeimportanttokeepinmindforthe developmentofnewtherapeutic approachestargeting theDNA damagesignalingandrepairpathways.

Conflictofintereststatement

Theauthorsdeclarethattherearenoconflictsofinterest.

Acknowledgements

Microscopic imaging wasperformed with equipment main- tainedby theSciencefor LifeLabBioVisPlatform,Uppsala.We sincerelythankK.KarlssonforhelpwithdatafortheM059cells.

AppendixA. Supplementarydata

Supplementarydataassociatedwiththisarticlecanbefound, in the online version, at http://dx.doi.org/10.1016/j.mrfmmm.

2014.06.004.

References

[1]P.L.Olive,TheroleofDNAsingle-anddouble-strandbreaksincellkillingby ionizingradiation,Radiat.Res.150(5Suppl.)(1998)S42–S51.

[2]A.Kakarougkas,P.A.Jeggo,DNADSBrepairpathwaychoice:anorchestrated handovermechanism,Br.J.Radiol.87(1035)(2014)20130685.

[3]A.Gospodinov,Z.Herceg,Chromatinstructureindoublestrandbreakrepair, DNARepair12(10)(2013)800–810.

[4]A.J.Davis,D.J.Chen,DNAdoublestrandbreakrepairvianon-homologousend- joining,Transl.CancerRes.2(3)(2013)130–143.

[5]A.A.Goodarzi,P.A.Jeggo,TherepairandsignalingresponsestoDNAdouble- strandbreaks,Adv.Genet.82(2013)1–45.

[6]R.T. Abraham, PI3-kinaserelated kinases: ‘big’players instress-induced signalingpathways,DNARepair3(8–9)(2004)883–887.

[7]E.J.Gapud,B.P.Sleckman,UniqueandredundantfunctionsofATMandDNA- PKcsduringV(D)Jrecombination,CellCycle10(12)(2011)1928–1935.

[8]S.P. Lees-Miller, K.Meek, Repairof DNA double strand breaks by non- homologousendjoining,Biochimie85(11)(2003)1161–1173.

[9]M.Shrivastav,L.P.DeHaro,J.A.Nickoloff,RegulationofDNAdouble-strand breakrepairpathwaychoice,CellRes.18(1)(2008)134–147.

[10]D.Deckbar,P.A.Jeggo,M.Lobrich,Understandingthelimitationsofradiation- inducedcellcyclecheckpoints,Crit.Rev.Biochem.Mol.Biol.46(4)(2011) 271–283.

[11]K.D.Mills,D.O.Ferguson,F.W.Alt,TheroleofDNAbreaksingenomicinstability andtumorigenesis,Immunol.Rev.194(2003)77–95.

[12]S.Burma,B.P.Chen,D.J.Chen,Roleofnon-homologousendjoining(NHEJ)in maintaininggenomicintegrity,DNARepair5(9–10)(2006)1042–1048.

[13]D.Huhn,H.A.Bolck,A.A.Sartori,TargetingDNAdouble-strandbreaksignalling andrepair:recentadvancesincancertherapy,SwissMed.Wkly.143(2013) w13837.

[14]F.M.Hsu,S.Zhang,B.P.Chen,RoleofDNA-dependentproteinkinasecatalytic subunitincancerdevelopmentandtreatment,Transl.CancerRes.1(1)(2012) 22–34.

[15]D.H.Auckley,R.E.Crowell,E.R.Heaphy,C.A.Stidley,J.F.Lechner,F.D.Gilliland, S.A.Belinsky,ReducedDNA-dependentproteinkinaseactivityisassociated withlungcancer,Carcinogenesis22(5)(2001)723–727.

[16]M.Someya,K.Sakata,Y.Matsumoto,H.Yamamoto,M.Monobe,H.Ikeda,K.

Ando,Y.Hosoi,N.Suzuki,M.Hareyama,TheassociationofDNA-dependent proteinkinaseactivitywithchromosomalinstabilityandriskofcancer,Car- cinogenesis27(1)(2006)117–122.

[17]B.Rigas,S.Borgo,A.Elhosseiny,V.Balatsos,Z.Manika,H.Shinya,N.Kurihara, M.Go,M.Lipkin,DecreasedexpressionofDNA-dependentproteinkinase,a DNArepairprotein,duringhumancoloncarcinogenesis,CancerRes.61(23) (2001)8381–8384.

[18]L.B.Smilenov,H.B.Lieberman,S.A.Mitchell,R.A.Baker,K.M.Hopkins,E.J.Hall, CombinedhaploinsufficiencyforATMandRAD9asafactorincelltransfor- mation,apoptosis,andDNAlesionrepairdynamics,CancerRes.65(3)(2005) 933–938.

[19]N.Tonotsuka,Y.Hosoi,S.Miyazaki,G.Miyata,K.Sugawara,T.Mori,N.Ouchi,S.

Satomi,Y.Matsumoto,K.Nakagawa,etal.,HeterogeneousexpressionofDNA- dependentproteinkinaseinesophagealcancerandnormalepithelium,Int.J.

Mol.Med.18(3)(2006)441–447.

[20]S.J.Arlander,B.T.Greene,C.L.Innes,R.S.Paules,DNAproteinkinase-dependent G2checkpointrevealedfollowingknockdownofataxia-telangiectasiamutated inhumanmammaryepithelialcells,CancerRes.68(1)(2008)89–97.

[21]Z.F.Shang,B.Huang,Q.Z.Xu,S.M.Zhang,R.Fan,X.D.Liu,Y.Wang,P.K.Zhou, InactivationofDNA-dependentproteinkinaseleadstospindledisruption andmitoticcatastrophewith attenuatedcheckpointprotein2phosphor- ylation in response to DNA damage, Cancer Res. 70 (9) (2010) 3657–

3666.

[22]B.Huang,Z.F.Shang,B.Li,Y.Wang,X.D.Liu,S.M.Zhang,H.Guan,W.Q.Rang, J.A.Hu,P.K.Zhou,DNA-PKcsassociateswithPLK1andisinvolvedinproper chromosomesegregationandcytokinesis,J.Cell.Biochem.115(6)(2014) 1077–1088.

[23]K.J.Lee,Y.F.Lin,H.Y.Chou,H.Yajima,K.R.Fattah,S.C.Lee,B.P.Chen,Involvement ofDNA-dependentproteinkinaseinnormalcellcycleprogressionthrough mitosis,J.Biol.Chem.286(14)(2011)12796–12802.

[24]S.P.Lees-Miller,R.Godbout,D.W.Chan,M.Weinfeld,R.S.Day,G.M.Barron3rd, J.Allalunis-Turner,Absenceofp350subunitofDNA-activatedproteinkinase fromaradiosensitivehumancellline,Science(NewYork,NY)267(5201)(1995) 1183–1185.

[25]S.J.DiBiase,Z.C.Zeng,R. Chen,T.Hyslop,W.J.CurranJr.,G.Iliakis,DNA- dependentproteinkinasestimulatesanindependentlyactive,nonhomologous, end-joiningapparatus,CancerRes.60(5)(2000)1245–1253.

[26]E.S.Williams,R.Klingler,B.Ponnaiya,T.Hardt,E.Schrock,S.P.Lees-Miller,K.

Meek,R.L.Ullrich,S.M.Bailey,TelomeredysfunctionandDNA-PKcsdeficiency:

characterizationandconsequence,CancerRes.69(5)(2009)2100–2107.

[27]B.Stenerlow,K.H.Karlsson,B.Cooper,B.Rydberg,MeasurementofpromptDNA double-strandbreaksinmammaliancellswithoutincludingheat-labilesites:

resultsforcellsdeficientinnonhomologousendjoining,Radiat.Res.159(4) (2003)502–510.

[28]L.Bozulic,B.Surucu,D.Hynx,B.A.Hemmings,PKBalpha/Akt1actsdownstream ofDNA-PKintheDNAdouble-strandbreakresponseandpromotessurvival, Mol.Cell30(2)(2008)203–213.

[29]A.M.Dragoi,X.Fu,S.Ivanov,P.Zhang,L.Sheng,D.Wu,G.C.Li,W.M.Chu,DNA- PKcs,butnotTLR9,isrequiredforactivationofAktbyCpG-DNA,EMBOJ.24 (4)(2005)779–789.

[30]B.P.Chen,N.Uematsu,J.Kobayashi,Y.Lerenthal,A.Krempler,H.Yajima,M.

Lobrich,Y.Shiloh,D.J.Chen,Ataxiatelangiectasiamutated(ATM)isessentialfor DNA-PKcsphosphorylationsattheThr-2609clusteruponDNAdoublestrand break,J.Biol.Chem.282(9)(2007)6582–6587.

[31]B.P.Chen,D.W.Chan,J.Kobayashi,S.Burma,A.Asaithamby,K.Morotomi-Yano, E.Botvinick,J.Qin,D.J.Chen,CellcycledependenceofDNA-dependentprotein kinasephosphorylationinresponsetoDNAdoublestrandbreaks,J.Biol.Chem.

280(15)(2005)14709–14715.

[32]S.W.Lee,K.J.Cho,J.H.Park,S.Y.Kim,S.Y.Nam,B.J.Lee,S.B.Kim,S.H.Choi,J.H.

Kim,S.D.Ahn,etal.,ExpressionsofKu70andDNA-PKcsasprognosticindicators oflocalcontrolinnasopharyngealcarcinoma,Int.J.Radiat.Oncol.Biol.Phys.

62(5)(2005)1451–1457.

[33]C.W.Anderson,T.H.Carter,TheDNA-activatedproteinkinase—DNA-PK,Curr.

Top.Microbiol.Immunol.217(1996)91–111.

[34]S.Zhang,P.Hemmerich,F.Grosse,CentrosomallocalizationofDNAdamage checkpointproteins,J.Cell.Biochem.101(2)(2007)451–465.

[35]D.W.Chan,B.P.Chen,S.Prithivirajsingh,A.Kurimasa,M.D.Story,J.Qin,D.J.

Chen,AutophosphorylationoftheDNA-dependentproteinkinasecatalytic subunitisrequiredforrejoiningofDNAdouble-strandbreaks,GenesDev.16 (18)(2002)2333–2338.

[36]B.B.Zhou,S.J.Elledge,TheDNAdamageresponse:puttingcheckpointsinper- spective,Nature408(6811)(2000)433–439.

References

Related documents

We have investigated the formation and repair of complex lesions, including double strand breaks (DSB) and clustered damages (two or more lesions within 10-20 base pairs) after

1) To investigate the influence of oxidative stress on the mtDNA replication replisome and the potential role of PrimPol as a translesion synthesis

Considering the important role of DNA-PKcs in DSB repair, any agent that can inhibit this protein or knock-down its related gene can be used to enhance the sensitivity of tumor

To study the ability of the cells to repair DNA double-strand breaks (DSBs) while they are depleted or deficient of key protein DNA protein kinase catalytic

Also the importance of chemically and ionizing radiation induced HLS repair and transformation into DSBs in live cells was assessed in DSB repair inhibited cells. The activation

DNA damaging agents like radiation and chemotherapeutic drugs are used in cancer treatment to induce DNA damage and cell death in tumor cells... Measurement of

Results from paper III demonstrated that repeated treatment using the isothiocyanate sulforaphane protected human skin fibroblasts from toxic effects of

This thesis investigates the role of two epigenetic players, miRNAs and DNA methylation, as well as the involvement of exosomes in paediatric glioma stem cells. The first study