/
REVIEWOpenAccess REVIEWOpenAccess

REVIEWOpenAccess - PDF document

alexa-scheidler
alexa-scheidler . @alexa-scheidler
Follow
366 views
Uploaded On 2017-11-27

REVIEWOpenAccess - PPT Presentation

Mousemodelsofsarcomascriticaltoolsinour understandingofthepathobiology SeanMPost Abstract SarcomasareneoplasticmalignanciesthattypicallyariseintissuesofmesenchymaloriginTheidentificationof novelmole ID: 610270

Share:

Link:

Embed:

Download Presentation from below link

Download Pdf The PPT/PDF document "REVIEWOpenAccess" is the property of its rightful owner. Permission is granted to download and print the materials on this web site for personal, non-commercial use only, and to display it on your personal computer provided you do not modify the materials and that you retain all copyright notices contained in the materials. By downloading content from our website, you accept the terms of this agreement.


Presentation Transcript

REVIEWOpenAccess Mousemodelsofsarcomas:criticaltoolsinour understandingofthepathobiology SeanMPost Abstract Sarcomasareneoplasticmalignanciesthattypicallyariseintissuesofmesenchymalorigin.Theidentificationof novelmolecularmechanismsleadingtosarcomaformationandtheestablishmentofnewtherapieshasbeen hamperedbyseveralcriticalfactors.First,thistypeofcancerisrarelyobservedintheclinicwithfewerthan15,000 newlycasesdiagnosedeachyearintheUnitedStates.Anothercomplicatingfactoristhatsarcomasareextremely heterogeneousastheyariseinamultitudeoftissuesfrommanydifferentcelllineages(e.g.bone(osteosarcoma), fat(liposarcoma),andmuscle(myosarcoma)).Thescarcityofclinicalsamplescoupledwithitsinherent heterogeneitycreatesachallengingexperimentalenvironmentforcliniciansandscientists.Facedwiththese challenges,therehasbeenextremelylimitedadvancementintreatmentoptionsavailabletopatientsascompared toothercancers.Inordertogleaninsightintothepathobiologyofsarcomas,scientistsarenowusing invivo mousemodelswhosegenomeshavebeenspecificallytailoredtocarrygenedeletions,geneamplifications,and pointmutationscommonlyobservedinhumansarcomas.Theuseofthesemodelorganismshasbeensuccessful inincreasingourknowledgeandunderstandingofhowalterationsinrelevantoncogenic,tumorsuppressive,and signalingpathwaysdirectlyimpactsarcomagenesis.Itisthegoalofmanyinthebiologicalcommunitythattheuse ofthesemousemodelswillserveaspowerful invivo toolstofurtherourunderstandingofsarcomagenesisand potentiallyidentifynewtherapeuticstrategies. Keywords: Sarcoma,Mousemodels,p53,Retinoblastoma(Rb),Translocation Background Sarcomasarearareformofcancerwithlessthan15,000 newcasesdiagnosedeachyearintheUnitedStates. Thoughrare,sarcomasarehighlydebilitatingmalignan- ciesastheyareoftenassociatedwithsignificantmorbidity andmortality.Sarcomasarebiologicallyveryheteroge- fromaplethoraofdifferenttissuesandcelltypes.They areclassicallydefinedbytheirtissueoforiginandareadd- itionallystratifiedbytheirhistopathologyorpatient ’ sage atdiagnosis[1,2].Whiletheseclassificationshaveproven useful,modernbiologicalandclinicaltechniqueshavethe abilitytofurtherstratifysarcomasbasedontheirgenetic profile[1,3,4].Cytogeneticandkaryotypeanalyseshave revealedtwodivergentgeneticprofilesinsarcomas.The firstandmostsimplegeneticprofileistheobservationof translocationeventsinsarcomaswithanotherwise normaldiploidkaryotype.Ontheotherhand,mostsarco- masdisplayamorecomplexgeneticphenotype,suggest- inggenomicinstabilityplaysanimportantroleinmany sarcomas. Historicalperspective Muchofourcurrentknowledgeregardingsarcomabiol- ogyhasbeenascertainedthroughexperimentationusing highdoseirradiation,viralinfections, invitro cellline studies,andxenograftsmodels.Oneoftheearliestanimal studiesinvestigatedtheimpactoftheRoussarcomavirus onthedevelopmentofsofttissuesarcomas[5].Our knowledgeregardingradiation-inducedsarcomagenesis allyexposedtoradiumandanimalmodelssubjectedto highdoseradiationdevelopedsarcomas[6,7].Whilethe plightofthesepatientsandthesubsequentanimalexperi- mentsledtotheidentificationofacauseandeffectfor somesarcomas,theseobservationswereunabletoidentify themoleculareventsresponsibleforsarcomagenesis. Correspondence: spost@mdanderson.org DepartmentofLeukemia,M.D.AndersonCancerCenter,1515Holcombe Blvd,Houston,TX77030,USA CLINICAL SARCOMA RESEARC H ©2012Post;licenseeBioMedCentralLtd.ThisisanOpenAccessarticledistributedunderthetermsoftheCreativeCommons AttributionLicense(http://creativecommons.org/licenses/by/2.0),whichpermitsunrestricteduse,distribution,and reproductioninanymedium,providedtheoriginalworkisproperlycited. Post ClinicalSarcomaResearch 2012, 2 :20 http://www.clinicalsarcomaresearch.com/content/2/1/20 Tomoreaccuratelyinvestigatethegeneticandmo-lecularchangesmanifestedinsarcomas,scientistsbeganusingpatientderivedsarcomacelllines.Thesecelllineshavealsoaddedtoourunderstandingofthesarcomadiseaseprogressioninvivo,throughtheiruseinxeno-graftexperiments[8-10].Eventhoughthesecelllineexperimentshavegreatlyadvancedourunderstandingofsarcomas,theyhaveseverelimitations.First,patient-derivedcelllinesaretypicallyisolatedduringsurgicalre-sectionoflatestagetumors[11].Thus,thesecellshaveundergonenumerousgeneticalterations,complicatingourabilitytoidentifythecriticalprimaryandsecondarygeneticcausesofthesecancers.Second,celllinesiso-latedfromindividualspossessdiversegeneticback-groundsashumansharbormillionsofsinglenucleotidepolymorphiccombinations[12].Finally,someofthecur-rentlyavailablesarcomacelllineshavebeenpassagedformorethanageneration.Theimpactofcellcultureshockiswelldocumentedandnodoubtaltersthemuta-tionrateandgeneticstabilityofthesecelllines[13].Howeachoftheseimpactsanindividualtumororitsresponsetotherapyislargelyunknown.Asecondcomplicationarisesfromtheuseofimmuno-compromisedxenograftmousemodelstransplantedwithhumansarcomacells.Theseexperimentshavetheabilitytotestthetumorformingpotentialofaparticularcellline;however,theyfailtofaithfullyrecapitulatethetrueinvivoenvironmentofasarcomaastheylackafunc-tionalimmunesystem[14].Itiswidelyappreciatedthattheimmunesurveillancesystemplaysacriticalroleintumorprevention[15].Furthermore,stromalinteractionsbetweenthehostandtheinjectedcelllinesdiffersignifi-cantlyandundoubtedlyalternormalmicroenvironmentinteractions.Giventhesecaveats,ithasbecomeimperativethatresearchersgeneratemoreaccurateanimalmodelsthatwillallowscientiststodirectlyinvestigatethemechan-ismsofsarcomagenesis.Inthisreview,wewillhighlightseveralmodelsengineeredtoharborknowntransloca-tionsthoughttodrivehumansarcomagenesisaswellastumorpronemodelswithanincreasedpropensityforsarcomaformation.Whilethisreviewisnotmeanttobecomprehensiveofallsarcomamodels,wewilldiscusshowspecificgeneticalterations,pathways,andanimalmodelsmayserveaspreclinicalmodelsforfuturestud-ies,andthusprovideaframeworkforotherstudiesexaminingtheimpactoftranslocationsorderegulatedpathways.SarcomasdefinedbytranslocationAsalludedtoabove,somesarcomasharbordiploidkaryo-typesbutposseschromosomaltranslocation,suggestingadirectcorrelationbetweenthetranslocationeventandtheetiologyofthedisease[16].Thespecificityofindividualtranslocationsarelikewiseusefuldiagnosticindicatorsofspecificsarcomas.Ewing'ssarcomascommonlycarryat(11;22)(q24:q12)reciprocaltranslocationresultinginagenefusionproductbetweentheRNAbindingpro-teinEwsandthetranscriptionfactorFli1[17,18].Giventhattherearefewerthan300newEwingssarcomacasesintheUnitedStateseachyear,ourunderstandingofthediseaseprocessisquitelimited.Therefore,inordertodirectlyinterrogatetheimpactofthefusiongeneontumorformation,severallaboratorieshavegeneratedmousemodelsexpressingantransgene.Alveolarrhabdomyosarcomas,likeEwingssarcomas,arealsooftendefinedbythepresenceoftranslocationevents,mostcommonlyt(2;13)(q35;q14)andt(1;13)(p36;q14)[19,20].However,themajorityofthesearethet(2;13)(q35;q14)translocationwhichresultsinthefusionofthetranscriptionfactorPax3withthetrans-activationdomainof[21].LikeEwingssarcoma,alveolarrhabdomyosarcomasareexceedinglyrare,withfewerthan100newcasesayearreportedintheUnitedStates.Sinceclinicalsamplesaredifficulttoobtain,ourknowledgeofthisdiseaseisquitesparse.Tocombatthisdilemma,severalmousemodelsmimickingthealveolarrhabdomyosarcomatranslocationeventshaverecentlybeengenerated.ThegenerationandcharacterizationofthealveolarrhabdomyosarcomaandEwingssarcomamousemodelsandtheirimpactontumorformationwillbedetailedinlatersections.SarcomaswithcomplexkaryotypesIncontrasttosarcomasidentifiedashavingdiploidkar-yotypes,themajorityofsarcomasbelongtothemorekaryotypicallycomplexgroup.Cytogeneticandkaryo-typicanalysesofundifferentiatedpleomorphicsarcomas,pleomorphicrhabdomyosarcomas,embryonalrhabdo-myosarcomas,andosteosarcomashaverevealedtheirgenomestobeunstableanddisorganizedasevidencedbymultipledeletions,amplifications,andchromosomalfusions[22].Molecularanalyseshaveshownthatmanyofthecanonicaltumorsuppressorpathways,suchasthep53andretinoblastomapathwaysareablatedinthesetumors[22].Furthermore,somesarcomasalsoharboractivatingoncogenicmutations;suchasexpres-sionofoncogenicK-ras.Together,disruptionofthesegenesandpathwaysarethoughttobeadrivingforceinsarcomagenesis.Unlikethedirectcorrelationbetweenasinglechromo-somaltranslocationeventindiploidsarcomas,itismorechallengingtoidentifywhichofthenumerousmuta-tions,deletions,oramplificationsdrivethedevelopmentofsarcomaswithcomplexcytogenetics.Furthermore,theextremeheterogeneityinthesesarcomasisalsoachallengeforcliniciansattemptingtodeveloppersona-lizedtreatmentstrategies.Giventhesecomplexities,weClinicalSarcomaResearchPage2of9http://www.clinicalsarcomaresearch.com/content/2/1/20 willhighlightsomeofthecriticalpathwaysthoughttobealteredduringsarcomagenesisbelow.TumorsuppressorandoncogenicpathwaysinvolvedinsarcomagenesisThep53pathwayThep53tumorsuppressorpathwayisoneofthemostwellcharacterizedpathwaysincancers[23].Thegeneencodesatranscriptionfactorrequiredfortheacti-vationofnumerousDNAdamage-dependentcheckpointresponseandapoptoticgenes[24,25],andthusitsactiv-itiesareoftenablatedinmanycancers.Inadditiontolossofp53functionsviainheritedgermlinemutations,thep53pathwayiscommonlydisruptedbypointmutationsinthegeneduringsporadicsarcomagenesis[26].However,eventhoughgenealterationsarewidelyregardedashavingasignificantimpactonsarcomagen-esis,manysarcomasretainwildtype,yetphenotypic-allydisplayalossofp53function.Thesefindingssuggestthatchangesinothercomponentsofthep53pathway;suchasamplificationofMdm2,anegativeregulatorofthep53pathway,mayresultinp53inactivation[27,28].Furthermore,bothmiceandhumanswithelevatedlevelsofMdm2duetoahighfrequencysinglenucleotidepoly-morphismintheMdm2promoter(Mdm2SNP309)aremoresusceptibletosarcomaformation[29-31].Add-itionally,deletionorsilencingofArfArfhuman),aninhibitoroftheMdm2-p53axis,oftenresultsindevelopmentofsarcomas.Together,thesedataindi-catethatwhileinactivationofthep53pathwayisobservedinthevastmajorityofhumansarcomas,themechanismsleadingtodisruptionofthepathwaycanvarygreatly.TheretinoblastomapathwayTheretinoblastoma(Rb)pathwayrepresentsasecondmajortumorsuppressorpathwayderegulatedinmanysar-comas.Individualsinheritingagermlinetypicallydevelopcancersoftheeyeearlyinlife[32-34].However,inadditiontoretinalcancers,thesechildrenhaveasignificantlyhigherpropensitytodevelopsarcomasthanthegeneralpopulation[35].Whileinheritanceofagerm-linealterationsincreasessarcomarisk,therearealsonumerousexamplesofsporadicsarcomasharboringspon-mutationsanddeletions,particularlyosteosar-comasandrhabdomyosarcomas[36].Furthermore,Ikn4aanegativeregulatoroftheCDK-cyclincomplexesthatphos-phorylateandactivateRb,isoftendeletedinsarcomas[37,38].Together,thesefindingsillustratetheimportanceoftheRbpathwayinsarcomagenesis.OncogenicsignalingInadditiontolossoftumorsuppressorpathways,sarcoma-genesisisalsodrivenbyaberrantoncogenicsignaling.TheRassignalingpathwayinparticularisthoughttobealteredduringsarcomadevelopment[39].DeregulationoftheRaspathwayaberrantlystimulatescellularproliferation,whichinandofitselfimpingesonthep53andRbpathways,col-lectivelydemonstratingthesignificantcross-talkbetweenthesethreeseparatebutoverlappingpathways.Giventhenumeroussignalingpathwayspotentiallydis-ruptedinsarcomas,therehasbeenacriticalneedtointer-rogatehoweachofthesegenesanddivergentpathwaysimpactsarcomagenesisinaprospectivemanner.Sincethesestudiesarenearlyimpossibleinhumanpatients,scientistsandcliniciansarenowusingmicegeneticallytai-loredforsuchstudies(Table1).Below,wewillhighlightseveralwellcharacterizedgeneticallyengineeredmousemodelsharboringcommongeneticalterationsobservedinsarcomabiology.MousemodelsofsarcomasFormanyyears,mousemodelshaveservedaspowerfultoolsinourinterrogationofthemechanismsregulatinghumancancers.However,itwasnotuntiltheprevalenceofgeneticallymanipulablemousemodelsinthe1980and90sthatwebecamefullycapableofexaminingthedirectcausesofmanycancersinaninvivosetting.Al-thoughwedonotfullyunderstandthediseaseprocessesofsarcomagenesis,wenowhaveamplebiologicalreagentsinwhichtoexploretheseprocesses,severalofwhicharedetailedbelow.MousemodelsharboringtranslocationssSarcomaSarcomaswithsimplediploidkaryotypesoftenhavechromosomaltranslocationsthatdirectlyimpactsarcoma-genesis.ToidentifytheimpactoftheEws-Fli1transloca-tion,t(11;22)(q24:q12),inEwingssarcoma,miceharboringtransgenehavebeengenerated.Expressionoftransgeneislethalwhenexpressedinsometis-sues[57].Therefore,tolimitthislethalphenotype,theEws-transgenemustbeconditionallyexpressedinspecificcelltypesusingtheCre-recombinase-loxPsystem[58].Cre-loxPtechnologieshavetheabilitytodeleteentiregenes,specificexons,orevenremoveinhibitorsoftrans-genicexpressioninspecificcelllineagesortissues[59].Usingthissystem,transgenicmiceharboringalatentEws-transgeneweregeneratedandcrossedwithmiceexpressingCre-recombinaseunderthecontrolofthePrxpromoter[44],resultingintheactivationofthetransgenespecificallyinosteogenicmultipotentcells.Al-thoughthesemicedevelopedmultipleboneabnormalities,theyultimatelyfailedtoproducesarco-mas.Thisfindingsuggeststhatwhilethet(11;22)(q24:q12)translocationisacommoneventinEwingssarcoma,itis,byitself,unabletostimulateacancerphenotypewhichindicatesthatotheraccompanyingmutations(orhitsClinicalSarcomaResearchPage3of9http://www.clinicalsarcomaresearch.com/content/2/1/20 thegenome)arerequiredforfranktumorformation.Toaddressthis,miceexpressingthetransgenewerethencrossedtomiceharboringPrx-Cre-directeddeletion.Themicerapidlydevelopedpoorlydifferentiatedsarcomas(medianageof21weeks);whilemediateddeletionofp53aloneresultedinthedevelopmentofosteosarcomas(medianageof50weeks),demonstratingthecooperativeinteractionsbetweenEws-Fli1andp53insarcomas.AlveolarrhabdomyosarcomasAlveolarrhabdomyosarcomasareoftencharacterizedbyt(2;13)(q35;q14)translocations.Knock-inmiceharboringthet(2;13)(q35;q14)translocationhavebeengeneratedbyknockingthegeneintothePax-3locus,result-inginaPax-3-FkhrfusiongeneunderthecontroloftheendogenousPax-3promoter[49].SimilartotheCre;Ews-Fli1studies,thesemicedidnotdevelopsarco-mas,butdiddisplaynumerouscongenitaldefects,sug-gestingthePax3-Fkhrfusiongeneisimportantinnormalmurinedevelopmentbutrequiresadditionalgen-etichitsforsarcomadevelopment.Inordertogenerateamorerobustalveolarrhabdomyosarcomamodel,micespecificallyexpressingaPax3-FkhrtransgeneinthemuscleundertheinfluenceofMyf6-Cre-mediatedacti-vationweregenerated[50,51].Surprisingly,thesemice Table1MousemodelsofhumansarcomasTumortypeGenealteration(s)AgentusedSignificanceProposedkaryotypeReferenceSoftTissueSarcomaNDRoussarcomaViralinfectioninfluencessarcomagenesisUnknown[OsteosarcomaNDRadiationRadiationinfluences7osteosarcomaformationinrabbitsUnknown[Sporadic/Variedp53NoneLossofresultsinsarcomaformationComplex[Sporadic/VariedmutationNoneMutationsinp53resultsinsarcomaformationComplex[Sporadic/31Variedworw/oNoneMutationsinthep53pathwayresultinsarcomaformationComplex[PoorlyDifferentiatedworw/oPrx-CreEws-Fli1micefailtoinducesarcomagenesisintheabsenceofTranslocationbutcomplexwithp53/Rblossloss44]PoorlyDifferentiatedSarcoma/Osteosarcomaworw/oRbandp53deletionsPrx-CreEws-Fli1micefailtoinducesarcomagenesisbutreducetimeofonsetintheabsenceofTranslocationbutcomplexwithwith45]OsteosarcomaTax;p19ArfNoneExpressionofTaxintheabsenceofp19ArfresultsinosteosarcomaformationComplex[OsteosarcomaRbandp53-CreDeletionofcooperateintheboneleadingtoosteosarcomagenesisComplex[Osteosarcoma/Leiomyosarcomaandp107deletionshaploinsufficiencycoupledwithdeletionresultsinlowpenetrantSarcomaComplex[DevelopmentaldefectsPax-3-FkhrfusionNonePax3-FkhrfusionproductfailstoproducesarcomaswhenexpressedfromPax3promoterTranslocation[RhabdomyosarcomasPax3-Fkhrw/andw/oMyf6-CreExpressionofthePax3-FkhrtransgenerequireslossofrhabdomyosarcomaformationTranslocationbutcomplexcomplex50,51]Rhabdomyosarcomas/VarioussarcomasPtch1heterozygosityw/andw/oMyf6-Cre,Myf-5Cre,Pax7-CreERLossofinconjunctionwithhaploinsufficiencyresultsinrhabdomyosarcomagenesisComplex[UndifferentiatedPleomorphicMutantK-rasexpressionandAdenoviralCreMutantK-rasexpressionandcooperateinthedevelopmentofundifferentiatedpleomorphicsarcomasComplex[PleomorphicRhabdomyosarcomasMutantK-rasexpressionandofCreintotheMutantK-rasexpressionandcooperateinthedevelopmentofundifferentiatedpleomorphicsarcomasComplex[RhabdomyosarcomasMutantK-rasexpressionandlossormutationAh-CreExpressionofmutantp53facilitatesamorerapiddevelopmentofrhabdomyosarcomasthanlossofinthemutantK-rasbackgroundComplex[ClinicalSarcomaResearchPage4of9http://www.clinicalsarcomaresearch.com/content/2/1/20 alsofailedtodisplayasarcomaphenotype.However,concomitantdeletionofArf,orintheMyf6-Cre;Pax3-Fkhrmiceresultedinarhabdomyosar-comaphenotype[50,51].Thesestudiesillustratethecomplexitiesinalveolarrhabdomyosacromagenesisandimplicatethep53andRbpathwaysinthedevelopmentofPax3-Fkhr-dependentsarcomas.AdditionalsarcomamousemodelsregulatedbytransloactioneventsSynovialsarcomas/myxoidliposarcomasTheidentificationofcommontranslocationeventshasgreatlyassistedinourunderstandingofsarcomagenesisandhasledtothegenerationofmousemodelswiththepowertoexaminetheirimpact.Inadditiontothetrans-locationsnotedabove,chromosomalrearrangementst(X;18)andt(12;16)(q12;p11)arecommonlyobservedinsynovialandliposarcomas,respectively(Table2).Mousemodelsmimickingthet(X;18)translocation,viaexpres-sionofthechimericproteinSYT-SSX2,resultinsyn-ovialsarcomaswithhighpenetrance[60,61].Likewise,expressionofTLS-CHOP,afusionproteinthatmimicsthet(12;16)(q12;p11)translocation,resultedinmyxoidroundcellliposarcomas[62].Giventherarenatureofthesetumors,thesemousemodelsmakeexcellentplat-formsforinvestigatingthepathobiologyofthesedis-easesaswellaspre-clinicaltherapeuticmodels[76,77].SarcomamousemodelswithcomplexgeneticsSarcomasofthebone(osteosarcomas)Incontrasttothesarcomasdrivenprimarilybyspecifictranslocations,themajorityofsarcomaspossesshighlyaneuploidgenomesduetodisruptionsintumorsuppres-sorpathwaysandaberrantoncogenicactivation.Osteo-sarcomasareoneofthemostwellstudiedtypesofsarcomaswithcomplexgeneticsgiventhedevelopmentofnumerousknock-out,knock-in,andtransgenicanimalmodelsavailableforthisdisease.Thegenerationandcharacterizationoftumorsfrom-nullandheterozygousknock-outmicedemonstratedtheimport-anceofp53inosteosarcomas[40,41].Theroleofp53inosteosarcomasisfurtherhighlightedbytumorana-lysisofknock-inmicecontainingamutantcopyofp53R172H(correspondingtotheR175Hhot-spotmuta-tioninhumans)[42,43].Animportantdifferentiationbetweentheknock-outandknock-inmiceisthatsarcomasdevelopedametastaticgainoffunctionphenotype,faithfullyrecapitulatingthepheno-typeobservedinthehumandisease[42,43].Thegener-ationofthemutantR172Hmousemodelprovidesresearchers,forthefirsttime,withtheabilitytoinvesti-gatemetastaticosteosarcomadiseaseprogressioninainvivosetting.Inadditiontodirectablationofp53function,transgenicmiceoverexpressingthep53regula-tor,Mdm2,aswellasmiceharboringasinglenucleotidepolymorphismintheMdm2promoter,haveanincreasedrisktodevelopsarcomas[31,68].Furthermore,trans-genicmiceexpressingtheviraloncogeneTax,coupledwithdeletionofArf,developedhighlypenetrantos-teosarcomas[46].Together,theseresultsfurtherdem-onstratetheimportanceofablatingthep53pathwayinosteosarcomagenesis.Inhumans,lossoftheRbpathwayhasalsobeenimplicatedintheetiologyofosteosarcomas.However,inthemouse,homozygousdeletionofresultsinanembryolethalphenotypeduetoplacentaldefects[69].Therefore,inordertoinvestigatetheroleofRbinbonemalignancies,researchersagainemployedtheCre-loxPsystemtodeletespecificallyinthebone.UnlikethecriticalroleofRbinhumanosteosarcomas,micelackinginosteocytesdonotdevelopcancers[47].However,whencoupledwithloss,lossexacerbatesthep53-dependentosteosarcomaphenotype,withmostmicesuc-cumbingtotheirdiseasewithin150days[45,47].Asacaveattothefindingthat-lossalonedidnotinduceosteosarcomas,thereissignificantredundancyintheRb Table2AdditionalmousemodelsofhumansarcomasTumortypeGenealteration(s)AgentusedSignificanceProposedkaryotypeReferenceSynovialSarcomaSYT-SSX2fusionMyf5-CreExpressionoftheSYT-SSX2transgeneresultedin100%penetrantsynovialsarcomasTranslocation[MyxoidLiposarcomaTLS-CHOPfusionw/p53deletionPrx-CreDeletionofcooperatesintheformationofliposarcomasTranslocation[NeurofibromaMPNSTworw/op19Arf3.9Periostin-CreDeletionofandp19ArfstimulateMPNSTdevelopmentComplex[UterineleiomyosarcomaLmp2deletionNoneLossofLmp2resultsinuterineleiomyosarcomaformationComplex[UterineleiomyosarcomaTDGF1/CRIPTOoverexpressionMMTV-promoterTDGF1/CRIPTOexpressionresultsinuterineleiomyosarcomadevelopmentComplex[ClinicalSarcomaResearchPage5of9http://www.clinicalsarcomaresearch.com/content/2/1/20 pathwayinmice.Rbconsistsofthreefamilymembers(p105,p107,andp130)andeachsharessimilarstructureandfunction[70].Assuch,concomitantlossofbothp107inmousedidinfactresultinalowpenetrantosteosarcomaphenotype[48,71].Takentogether,thesestudiesdemonstratetheabsoluterequirementforablationofthep53pathwayinosteosarcomagenesisandsuggestthatpRbplaysaco-operativeroleinosteosarcomagenesis.SofttissuesarcomasUndifferentiatedpleomorphicsarcomasUndifferentiatedpleomorphicsarcomasaresofttissuesarcomastypicallyobservedinadultsthatarisefromcellsofunknownorigin,and,likeosteosarcomas,displaycomplexgeneticsresultingfromderegulationofmultiplepathways.Investigationsintothecellularoriginofbothundifferentiatedpleomorphicsarcomasandembryonalrhabdomyosarcomashaveidentifiedtheimportanceofthep53andRbpathwaysintheetiologyofbothmalig-nancies[52].Inadditiontotheimportanceofthesetwotumorsuppressorpathways,theKras-signalingpathwayhasalsobeenimplicatedinthedevelopmentofundiffer-entiatedpleomorphicsarcomas[53,54].MiceharboringalatentcopyofoncogenicKrasLSLG12D(silencedbyafloxedloxP-stop-loxP(LSL)cassette)andtwofloxed)thatweresimultaneouslyactivatedtoexpressmutantanddeletefollowinginjectionofadenoviral-Creintothemuscle,rapidlydevelopedsarco-maswithsignificantmetastaticpotential.Detailedmolec-ularanalysisoftheAd-cre;KrasG12Dtumorsrevealedanexpressionprofilesimilartothoseobservedinhumanundifferentiatedpleomorphicsarcomas[54].Together,thesedatasupporttheideathatbothablationoftumorsuppressorpathwaysandactivationofoncogenescooper-atetodrivesarcomagenesis.RhabdomyosarcomasUsingtheCre-LoxPstrategytosimultaneouslyactivateala-tentoncogenicK-rasG12Valleleanddeletethe2-10allelesinmyocytes,itwasdemonstratedthatmicerapidlydevelopsarcomasthatarehistopathologicallysimilartopleomorphicrhabdomyosarcomasobservedinhumans[55].AlthoughtheundifferentiatedpleomorphicandrhabdomyosarcomastudiesusedsimilarmousemodelstoidentifytheroleofmutantK-rasand-lossinsarcoma-genesis,theseexperimentsresultedinsomewhatdifferentmalignancies.Thus,giventhecellularsimilaritiesbetweenundifferentiatedpleomorphicsarcomasandrhabdomyo-sarcomas[52],itisimperativetofurtherinvestigatesarco-magenesisintheKras-LSLG12DFl210/Fl2mousemodelsusingmultiplemyospecificCre-expressingtrans-genicmiceinordertopreciselyascertainhowthesepath-wayssynergiesinspecifictissues.WhileeachoftheKras-LSL10/Fl2studiesmentionedaboverevealtheimportanceofp53andK-rasinmyocytespecificsarcomagenesis,theyfailedtoac-curatelyrepresentthemostcommontypeofalterationtothegeneinhumancancers(e.g.p53mutations).Arecentstudyexaminedtheimpactofp53insarcoma-genesismoreaccuratelybynotonlydeletingalsoexpressingthemutant(correspondingtothehumanp53R175hotspotmutation)inthemuscle[56].UsingtheKrasLSLG12V;p53Fl210/Fl2KrasLSLG12VR172H/Fl2allelesincombinationwithAh-Creexpres-sion,itwasrevealedthatexpressionofmutantp53,eveninthecontextofheterozygosity(e.g.,p53R172H/+),hadamoredeleteriouseffectthansimplylosingonewildtypeallele.TheseAh-Cre;KrasG12VR172H/miceformedrhabdomyosarcomaswithhighpenetranceascomparedtolessthan10%rhabdomyosarcomasformationintheCre;KrasG12Vmice.Inaddition,unlikethetumorsAh-Cre;KrasG12Vmice,thetumorsfromtheAh-Cre;KrasG12V;p53R172H/micealsorecapitulatedthemetastaticphenotypetypicallyobservedinhumanrhabdomyosarcomas.AdditionalsarcomamousemodelsregulatedbydrivermutationsNeurofibromatosis/leiomyosarcomasGiventheextremeheterogeneityofsarcomaswithregardstotissueoforigin,itisobviousthatalterationstonumerousgenes,pathways,andsignalingcomplexesplayanimportantroleinthepathobiologyofsarco-mas.Whilethisreviewdoesnotcoverallgeneticalterationsresponsibleforsarcomadevelopment,therearenumerousadditionalgenesthatimpactthisdisease(Table2).Forexample,alterationsinexpressionoftumorsuppressorgenes,suchasNeurofibromatosistype1(NF1),likewiseimpacttheetiologyofsomesar-comas.Mousemodelsthatcarrygenomicdeletionsand/ortissue-specificCre-mediateddeletionofresultinneurofibromas[72].TheseNF1-dependentphenotypesarefurtherexacerbatedwheniscon-comitantlydeletedwithothertumorsuppressors(e.g.;)resultinginmoreaggressivephenotypesasevidencedbymalignantperipheralnervesheathtumorformation[63,64].Tofurtherillus-tratethatlossofasinglegeneimpactssarcomaforma-tion,miceharboringanLMP-2deletionresultedinspontaneousuterineleiomyosarcomas[65].Thispro-videsevidenceofitsroleasatumorsuppressorandapotentialbiomarkerinhumandisease[66,73].Inadditiontolossoffunctionalterations,overexpressionofteratocarcinoma-derivedgrowthfactor1,alsoknownasCRIPTO,resultsinleiomyosarcomasbyde-regulationoftheWNTpathway[67].ClinicalSarcomaResearchPage6of9http://www.clinicalsarcomaresearch.com/content/2/1/20 Thevastdifferencesinthecellularoriginsofsarcomas,thelackofavailabilityoftumorspecimens,andthehetero-geneityinherentwithinindividualtumorshasimpededourabilitytofullyunderstandthebiologyofsarcomas.However,giventheavailabilityofnumerousgeneticknock-outs,knock-ins,andconditionalallelescoupledwiththebevyoftissue-specificCre-recombinaseexpres-singmouselines,wenowhavetheabilitytosystematicallyandprospectivelyinterrogatehowindividualgenesandmutationsimpactsarcomagenesis.Goingforward,tumoranalysisfrommultiplemurinederivedtumortypescanbecomparedandcontrastedinordertoidentifycriticalchangesinspecificsarcomas.Thesemousemodelshaveclearlydemonstratedthatwhiletherearedrivermuta-tions/translocations,sarcomagenesisis,infact,amulti-hitdisease.Theuseofthesemousemodelsmimickingthehumandiseaseconditionleadstoacriticalquestion:whattherapeuticapproachescanbetakentolessentheimpactofthesedebilitatingdiseases?First,wemustrecognizethatthesemousemodelsdemonstratethesynergismbetweenmultiplepathwaysandthuscombinatorialtreatmentstrat-egiesareneededtocombatthesecancers.Fortreatmentofpatientswithtranslocations,onecanenvisionatargetedtherapeuticapproach,likethatwhichhasbeenobservedinthetreatmentofchronicmyeloidleukemia.Theadditionoftyrosinekinaseinhibitors(TKIs),suchasima-tinib,whichinhibitstheactivityoftheBCR-ABLfusiongene,hasreducedCMLfromadeathsentencetoaman-ageableandstabledisease.Canthescientific/clinicalcom-munitydesigntargetdrugstothetranslocationeventsobservedinsarcomas?Theuseofthesemousemodelsmayserveasanexcellentpreclinicalplatformforsuchstudies.Treatingandalleviatingthediseaseprocessinsarcomaswithcomplexgeneticsmayprovemoredifficultthaniden-tifyingtargetedtherapies.However,giventhatmanygroupshaveidentifiedtheimportanceofspecificpathwaysinsarcomagenesis,suchasthep53pathway,wehaveastartingpoint.PreclinicaldrugslikePRIMA1-MetandNCS319726havebeenshowntorestoremutantp53activ-ities[74,75].Thesedrugscouldberapidlyscreenedforef-ficacyinmutantp53sarcomamodels.Moreover,thep53pathwayisalsoinactivatedbydysregulationofitsproteinpartners,Mdm2andp19Arf.TheemploymentofMdm2-p53antagonists,suchasNutlin-3andRITAmayproveef-ficaciousinreactivatingthep53pathwayandthusprovideatherapeuticbenefit.Also,lossofp19ARFduetopromotermethylationisacommoneventinsarcomagenesis.There-fore,theseanimalmodelsmayproveusefulinexaminingtheimpactofhypomethylatingagents,suchasazacytidineordasatinib,insarcomas.Incaseswherespecificoncogenesareknowntodrivetumorformation,suchasactivatedK-ras,theuseofcompoundsthatinhibitK-rastargets(suchasMEK)couldbebeneficial.TheefficacyofaMEK-inhibitorlikeARRY-162couldbereadilyexaminedinmousemodelspossessingamutatedK-rassignalingpathway.Allofthesepotentialchemotherapeuticagents,ifproveneffectiveininvivoclinicalmodels,couldprovidearationaleforpersonalizedandtargetedtherapyinsarcomapatients.Whilemousemodelscannotcompletelypredicttheout-comeofeachdisease,theycanprovidevaluableandcriticalinformation,particularlyinexceedinglyraretypesofsarco-masorwhenlowpenetrantsinglenucleotidepolymorph-ismsconfounddataanalysis.Rb:Retinoblastoma;Cre:Cre-recombinase.CompetinginterestsTheauthordeclarethathehavenocompetinginterests.SMPwrotethemanuscript.AcknowledgementsIwouldliketothankMs.XiaoruiZhangforreviewingthemanuscript.Received:27March2012Accepted:16July2012Published:4October20121.LasotaJ,Fanburg-SmithJC:Geneticsforthediagnosisandtreatmentofmesenchymaltumors.SeminMusculoskeletRadiol2.DavicioniE,etalMolecularclassificationofrhabdomyosarcomagenotypicandphenotypicdeterminantsofdiagnosis:areportfromtheChildren'sOncologyGroup.AmJPathol3.HelmanLJ,MeltzerP:Mechanismsofsarcomadevelopment.NatRev4.TaylorBS,etalAdvancesinsarcomagenomicsandnewtherapeuticNatRevCancer5.RousP:ASarcomaoftheFowlTransmissiblebyanAgentSeparablefromtheTumorCells.JExpMed6.MartlandHS:Theoccuranceofmalignancyinradioactivepersons.AmericanJournalofCancer7.SabinFR,DoanCA,ForknerCE:TheProductionofOsteogenicSarcomataandtheEffectsonLymphNodesandBoneMarrowofIntravenousInjectionsofRadiumChlorideandMesothoriuminRabbits.JExpMed8.BudachV,etalRadioresponsivenessofahumansofttissuesarcomaxenografttodifferentsingleandfractionatedregimens.Strahlenther9.KodousekR,etalHistopathologicalandultrastructuralobservationsofsomehumantumorxenograftspassagedinathymicnudemice.I.Humanosteosarcomaxenograftwithfeaturesofviral(oncornaAandCtyperetrovirus)involvement.ActaUnivPalackiOlomucFacMed10.PimmMV,BaldwinRW:Serologicalaspectsofrattumourxenograftgrowthinathymicnudemice.BrJCancer11.LinPP,etalSurgicalmanagementofsofttissuesarcomasofthehandandfoot.12.IstrailS,etalWhole-genomeshotgunassemblyandcomparisonofhumangenomeassemblies.ProcNatlAcadSciUSA13.SherrCJ,DePinhoRA:CellularSenescence:MinireviewMitoticClockorCultureShock?14.PelleitierM,MontplaisirS:Thenudemouse:amodelofdeficientT-cellMethodsAchievExpPathol15.WherryEJ:Tcellexhaustion.NatImmunol16.FletcherJA:Cytogeneticsandexperimentalmodelsofsarcomas.CurrOpinOncolClinicalSarcomaResearchPage7of9http://www.clinicalsarcomaresearch.com/content/2/1/20 17.BoninG,etalChimericEWS-FLI1transcriptinaEwingcelllinewithacomplext(11;22;14)translocation.CancerRes18.Turc-CarelC,etalChromosomaltranslocation(11;22)incelllinesofEwing'ssarcoma.CRSeancesAcadSciIII19.Turc-CarelC,etalConsistentchromosomaltranslocationinalveolarCancerGenetCytogenet20.BiegelJA,etalChromosomaltranslocationt(1;13)(p36;q14)inacaseofGenesChromosomesCancer21.BarrFG,etalRearrangementofthePAX3pairedboxgeneinthepaediatricsolidtumouralveolarrhabdomyosarcoma.NatGenet22.ToguchidaJ,NakayamaT:Moleculargeneticsofsarcomas:applicationstodiagnosesandtherapy.CancerSci23.VogelsteinB,LaneD,LevineAJ:Surfingthep53network.24.RaycroftL,etalAnalysisofp53mutantsfortranscriptionalactivity.MolCellBiol25.RaycroftL,WuHY,LozanoG:Transcriptionalactivationbywild-typebutnottransformingmutantsofthep53anti-oncogene.Science26.WangLL:Biologyofosteogenicsarcoma.CancerJ27.OlinerJD,etalAmplificationofageneencodingap53-associatedproteininhumansarcomas.28.OlinerJD,etalOncoproteinMDM2concealstheactivationdomainoftumoursuppressorp53.29.BondGL,etalASingleNucleotidePolymorphismintheMDM2PromoterAttenuatesthep53TumorSuppressorPathwayandAcceleratesTumorFormationinHumans.30.ItoM,etalComprehensiveMappingofp53PathwayAlterationsRevealsanApparentRoleforBothSNP309andMDM2AmplificationinClinCancerRes31.PostSM,etalAHigh-FrequencyRegulatoryPolymorphisminthep53PathwayAcceleratesTumorDevelopment.CancerCell32.TebbetRD,VickeryRD:Osteogenicsarcomafollowingirradiationforretinoblastoma;withthereportofacase.AmJOphthalmol33.NordlingCO:Anewtheoryoncancer-inducingmechanism.BrJCancer34.KnudsonAGJr:Mutationandcancer:statisticalstudyofretinoblastoma.ProcNatlAcadSciUSA35.DeshpandeA,HindsPW:Theretinoblastomaproteininosteoblastdifferentiationandosteosarcoma.CurrMolMed36.ToguchidaJ,etalPreferentialmutationofpaternallyderivedRBgeneastheinitialeventinsporadicosteosarcoma.37.YonghaoT,etalDeletionsandpointmutationsofp16,p15geneinprimarytumorsandtumorcelllines.ChinMedSciJ38.OdaY,etalFrequentalterationofp16(INK4a)/p14(ARF)andp53pathwaysintheroundcellcomponentofmyxoid/roundcellliposarcoma:p53genealterationsandreducedp14(ARF)expressionbothcorrelatewithpoorprognosis.JPathol2005,39.BourdeautF,etalMosaicismforoncogenicG12DKRASmutationassociatedwithepidermalnevus,polycystickidneysandJMedGenet40.JacksT,etalTumorspectrumanalysisinp53-mutantmice.CurrBiol41.DonehowerLA,etalMicedeficientforp53aredevelopmentallynormalbutsusceptibletospontaneoustumours.42.LangGA,etalGainoffunctionofap53hotspotmutationinamousemodelofLi-Fraumenisyndrome.43.OliveKP,etalMutantp53gainoffunctionintwomousemodelsofLi-Fraumenisyndrome.44.LinPP,etalEWS-FLI1inducesdevelopmentalabnormalitiesandacceleratessarcomaformationinatransgenicmousemodel.CancerRes45.LinPP,etalTargetedmutationofp53andRbinmesenchymalcellsofthelimbbudproducessarcomasinmice.46.RauchDA,etalTheARFTumorSuppressorRegulatesBoneRemodelingandOsteosarcomaDevelopmentinMice.PLoSOne47.WalkleyCR,etalConditionalmouseosteosarcoma,dependentonp53lossandpotentiatedbylossofRb,mimicsthehumandisease.GenesDev48.DannenbergJH,etalTissue-specifictumorsuppressoractivityofretinoblastomagenehomologsp107andp130.GenesDev49.LagutinaI,etalPax3-FKHRknock-inmiceshowdevelopmentalaberrationsbutdonotdeveloptumors.MolCellBiol50.KellerC,etalAlveolarrhabdomyosarcomasinconditionalPax3:Fkhrmice:cooperativityofInk4a/ARFandTrp53lossoffunction.GenesDev51.KellerC,etalPax3:FkhrinterfereswithembryonicPax3andPax7function:implicationsforalveolarrhabdomyosarcomacelloforigin.GenesDev52.RubinBP,etalEvidenceforanUnanticipatedRelationshipbetweenUndifferentiatedPleomorphicSarcomaandEmbryonalCancerCell53.KirschDG,etalAspatiallyandtemporallyrestrictedmousemodelofsofttissuesarcoma.NatMed54.MitoJK,etalCrossspeciesgenomicanalysisidentifiesamousemodelasundifferentiatedpleomorphicsarcoma/malignantfibrousPLoSOne55.TsumuraH,etalCooperationofoncogenicK-rasandp53deficiencyinpleomorphicrhabdomyosarcomadevelopmentinadultmice.56.DoyleB,etalp53mutationandlosshavedifferenteffectsontumourigenesisinanovelmousemodelofpleomorphicJPathol57.TorchiaEC,etalEWS/FLI-1inducesrapidonsetofmyeloid/erythroidleukemiainmice.MolCellBiol58.SauerB,HendersonN:Site-specificDNArecombinationinmammaliancellsbytheCrerecombinaseofbacteriophageP1.ProcNatlAcadSci59.LaksoM,etalTargetedoncogeneactivationbysite-specificrecombinationintransgenicmice.ProcNatlAcadSciUSA60.HaldarM,etalAconditionalmousemodelofsynovialsarcoma:insightsintoamyogenicorigin.CancerCell61.HaldarM,etalACreER-basedrandominductionstrategyformodelingtranslocation-associatedsarcomasinmice.CancerRes62.CharytonowiczE,etalPPARgammaagonistsenhanceET-743-inducedadipogenicdifferentiationinatransgenicmousemodelofmyxoidroundcellliposarcoma.JClinInvest63.BuchstallerJ,McKeeverPE,MorrisonSJ:TumorigeniccellsarecommoninmouseMPNSTsbuttheirfrequencydependsupontumorgenotypeandassayconditions.CancerCell64.JosephNM,etalThelossofNf1transientlypromotesself-renewalbutnottumorigenesisbyneuralcreststemcells.CancerCell65.HayashiT,FaustmanDL:Developmentofspontaneousuterinetumorsinlowmolecularmasspolypeptide-2knockoutmice.CancerRes66.HayashiT,etalMolecularApproachtoUterineLeiomyosarcoma:LMP2-DeficientMiceasanAnimalModelofSpontaneousUterine67.StrizziL,etalDevelopmentofleiomyosarcomaoftheuterusinMMTV-CR-1transgenicmice.JPathol68.JonesSN,etalOverexpressionofMdm2inmicerevealsap53-independentroleforMdm2intumorigenesis.ProcNatlAcadSciUSA69.deBruinA,etalRbfunctioninextraembryoniclineagessuppressesapoptosisintheCNSofRb-deficientmice.ProcNatlAcadSciUSA70.RizzolioF,etalRBgenefamily:genome-wideChIPapproachescouldopenundiscoveredroads.JCellBiochem71.Robanus-MaandagE,etalp107isasuppressorofretinoblastomadevelopmentinpRb-deficientmice.GenesDevClinicalSarcomaResearchPage8of9http://www.clinicalsarcomaresearch.com/content/2/1/20 72.BrossierNM,CarrollSL: Geneticallyengineeredmousemodelsshednew lightonthepathogenesisofneurofibromatosistypeI-relatedneoplasms oftheperipheralnervoussystem. BrainResBull 2012, 88 (1):58 – 71. 73.HayashiT, etal : PotentialroleofLMP2astumor-suppressordefinesnew targetsforuterineleiomyosarcomatherapy. SciRep 2011, 1: 180. 74.BykovVJN, etal : PRIMA-1METsynergizeswithcisplatintoinducetumor cellapoptosis. Oncogene 2005, 24 (21):3484 – 3491. 75.YuX, etal : Allele-Specificp53MutantReactivation. CancerCell 2012, 21 (5):614 – 625. 76.FrapolliR, etal : Novelmodelsofmyxoidliposarcomaxenografts mimickingthebiologicalandpharmacologicfeaturesofhumantumors. ClinCancerRes 2010, 16 (20):4958 – 4967. 77.JonesKB, etal : Ofmiceandmen:opportunitiestousegenetically engineeredmousemodelsofsynovialsarcomaforpreclinicalcancer therapeuticevaluation. CancerControl 2011, 18 (3):196 – 203. doi:10.1186/2045-3329-2-20 Citethisarticleas: Post: Mousemodelsofsarcomas:criticaltoolsinour understandingofthepathobiology. ClinicalSarcomaResearch 2012 2 :20. Submit your next manuscript to BioMed Central and take full advantage of: € Convenient online submission € Thorough peer review € No space constraints or color “gure charges € Immediate publication on acceptance € Inclusion in PubMed, CAS, Scopus and Google Scholar € Research which is freely available for redistribution Submit your manuscript at www.biomedcentral.com/submit Post ClinicalSarcomaResearch 2012, 2 :20 Page9of9 http://www.clinicalsarcomaresearch.com/content/2/1/20