配色: 字号:
Prox1 Promotes Expansion of theColorectalCancer Stem Cell Population to Fuel Tumor Growth and Ischem
2015-12-21 | 阅:  转:  |  分享 
  
Article

theColorectalCancerStem

andIschemia

Highlights

Prox1expressionisdispensableforhomeostasisinthenormal

intestine

AsubpopulationofProx1

+

cellshasstemcellactivityinintesti-

naladenomas/CRC

tumorcell

Authors

Zolta′nWiener,JennyHo¨gstro¨m,...,Yinon

Ben-Neriah,KariAlitalo

Correspondence

kari.alitalo@helsinki.fi

Wieneretal.nowshowthattheProx1

Wieneretal.,2014,CellReports8,1943–1956

September25,2014a2014TheAuthors

http://dx.doi.org/10.1016/j.celrep.2014.08.034

AnnexinA1andfilaminAmediateProx1effectsonstemcellac-

tivityinthetumors

LossofProx1decreasesadenoma/CRCstemcells,

growth,andsurvival

CellPopulationtoFuelTumorGrowth

Resistance

GraphicalAbstract

Prox1PromotesExpansionof

transcriptionfactorfunctionsasastem

cellregulatorinintestinaladenomasand

colorectalcancer(CRC),butnotinthe

normalintestine.Prox1criticallycontrib-

utestotumorcellsurvivalinhypoxia

andtotheexpansionoftheadenoma/

CRCstemcellpopulationviainhibition

oftheWnt-targetannexinA1andinduc-

tionoftheactin-bindingproteinfilamin

A.TheProx1pathwaythusrepresents

anattractivetherapeutictargetfordrug

developmentinCRC.

AccessionNumbers

GSE47568

InBrief

CellReports

Article

Prox1PromotesExpansion

oftheColorectalCancerStemCellPopulation

toFuelTumorGrowthandIschemiaResistance

Zolta′nWiener,

1,6

JennyHo¨gstro¨m,

1,6

VilleHyvo¨nen,

1

ArjaM.Band,

1

PauliinaKallio,

1

TanjaHolopainen,

1

OlliDufva,

1

CajHaglund,

3

OlliKruuna,

3

GuillermoOliver,

4

YinonBen-Neriah,

5

andKariAlitalo

1,2,



1

TranslationalCancerBiologyProgram,UniversityofHelsinki

2

WihuriResearchInstitute

BiomedicumHelsinki,00014Helsinki,Finland

3

DepartmentofSurgery,HelsinkiUniversityCentralHospital,00029Helsinki,Finland

4

DepartmentofGenetics,StJudeChildren’sResearchHospital,Memphis,TN38105,USA

5

LautenbergCenterforImmunology,HebrewUniversity-HadassahMedicalSchool,Jerusalem91120,Israel

6

Co-firstauthor

Correspondence:kari.alitalo@helsinki.fi

http://dx.doi.org/10.1016/j.celrep.2014.08.034

ThisisanopenaccessarticleundertheCCBYlicense(http://creativecommons.org/licenses/by/3.0/).

SUMMARY

Colorectalcancer(CRC)initiationandgrowthisoften

attributedtostemcells,yetlittleisknownaboutthe

regulationofthesecells.Weshowherethatasub-

populationofProx1-transcription-factor-expressing

cellshavestemcellactivityinintestinaladenomas,

butnotinthenormalintestine.Usinginvivomodels

and3Dexvivoorganoidculturesofmouseade-

nomasandhumanCRC,wefoundthatProx1dele-

tionreducedthenumberofstemcellsandcell

proliferationanddecreasedintestinaltumorgrowth

viainductionofannexinA1andreductionofthe

actin-bindingproteinfilaminA,whichhasbeenimpli-

catedasaprognosticmarkerinCRC.LossofProx1

alsodecreasedautophagyandthesurvivalof

hypoxictumorcellsintumortransplants.Thus,

Prox1isessentialfortheexpansionofthestemcell

poolinintestinaladenomasandCRCwithoutbeing

criticalforthenormalfunctionsofthegut.

INTRODUCTION

Colorectalcancer(CRC)isoneoftheleadingcausesofcancer

mortalityinWesterncountries.InmostCRCpatients,aninitial

mutationoccursintheAPCorCTNNB1gene,leadingtoactiva-

tionoftheb-catenin/TCF(canonicalWnt)pathway(Fearon,

2011;FoddeandSmits,2001).Inthecryptsofthenormalgut,

theb-catenin/TCFpathwayisactiveinPanethcells,intransit-

amplifying(TA)/progenitorcellsthathavealimitedproliferative

capacity,andinLgr5

+

intestinalstemcells(Clevers,2006).

Lgr5

+

cellsarecapableofefficientlyinitiatingadenomaformation

uponmutationoftheApcgene(Barkeretal.,2009).Inaddition,

progenitorsofintestinalepithelialcellscanconverttoastem

cell-likephenotypeandcontributetotheinitiationofCRCunder

inflammatoryconditions(Schwitallaetal.,2013).Thesestudies

indicatethattheacquisitionofastemcell-likephenotypeis

criticalforCRCtumorigenesis.

Intestinaladenomasarehighlyheterogenous,containingboth

proliferatinganddifferentiatingcells,andtheyarecontinuously

maintainedbyadedicatedcellpopulation,theso-calledcancer

stemcells(SampieriandFodde,2012;Schepersetal.,2012).

Althoughneoplasticcellsarecharacterizedbyincreasedcell

proliferationandlimitedcelldifferentiationcapacity,their

detaileddifferentiationpathwaysinCRCarepoorlyknown.

ExpressionoftheLgr5genehasbeenshowntomarkacellpop-

ulationwithstemcellpropertiesinmouseintestineandinintes-

tinaladenomas(Barkeretal.,2007;Schepersetal.,2012).

Furthermore,theintestinalstemcellsignature,includingLGR5

expression,identifiesCRCstemcellsandpredictsdisease

relapsealsoinhumanCRCpatients(Kemperetal.,2012;Mer-

los-Sua′rezetal.,2011).

AfterthegeneticlesionthatactivatestheWntsignaltransduc-

tionpathwayandabnormalcellproliferation,additionalmuta-

tionsaccumulateslowlytopromoteadenomaprogression

towardCRC,tumorinvasion,andmetastasis(Sampieriand

Fodde,2012).HighlyelevatedWntactivityafterApcdeletionin-

ducesexpressionofthehomeoboxtranscriptionfactorProx1in

intestinaltumorcells.WhenProx1wasdeletedinmicewithan

Apcmutation,adenomagrowthanddevelopmentofdysplasia

inthetumorepitheliumwasinhibited(Petrovaetal.,2008).In-

terestingly,Prox1upregulationafterlossofthetp53tumorsup-

pressorcontributestointestinaltumorprogressioninsome

modelsystems(Elyadaetal.,2011).

Here,weanalyzedthemechanismofProx1-inducedintestinal

adenomaprogressioninmicrosatellite-stabletumormodels.We

foundthatasubpopulationofProx1

+

cellsdisplaysstemcell

activityinadenomas/CRC,butnotinthenormalintestinal

epithelium.Furthermore,Prox1deletionreducedthesizeofthe

Lgr5

+

adenomaandCRCstemcellpopulations,andstemcell

activityandledtoreducedgrowthanddecreasedtumorcellsur-

vivalinanunfavorablemicroenvironment.

CellReports8,1943–1956,September25,2014a2014TheAuthors1943

RESULTS

Prox1ExpressionIsInducedinLgr5

+

CellsuponApc

GeneDeletion

TocharacterizeProx1-expressingcellsinthepathogenesisofin-

testinaladenomas,weinducedanacutedeletionoftheApc

genethroughoutthewholeintestinalepitheliuminApc

flox/flox

;

villin-Cre

ER

(VApc)micebyasingletamoxifeninjection

(VApcD/D).Asexpected,Panethcells,markingthecrypt

bottomsinthewild-type(WT)smallintestine,weredislocated

towardthelumeninmostoftheApc-deletedcrypts,andex-

pansionofthecellpopulationwithanactiveWntsignaling

pathwaywasdetectedbyEphB2immunostaining6daysafter

thetamoxifeninjection(FiguresS1AandS1B)(Batlleetal.,

2002).MostcryptscontainedscatteredProx1

+

cellclusters

intermingledwithlysozyme-positivePanethcells(FigureS1A).

CyclinD1markstheproliferatingcellpopulation,includingthe

progenitorcellsinWTintestine(GregorieffandClevers,2005).

Inlinewithpreviouslypublishedresults,thecyclinD1

+

cellpop-

ulationexpandedaftertheApcdeletion(Sansometal.,2005)

(FiguresS1AandS1B).CyclinD1

high

cellswerelocated

frequentlyclosetotheProx1

+

cells;however,Prox1

+

cells

werenotcyclinD1

high

inVApcD/Dmiceandintumorsfrom

Apc

min/+

mice(FiguresS1CandS1D)orfromCRCpatients(Fig-

ureS1E).Similarlytotheinvivoexperiments,whenweisolated

intestinalorganoidsfromtheVApcmiceandinducedApc

genedeletionin3DMatrigelculturebytheadditionof4-hy-

droxy-tamoxifen(4-OH-Tam),theorganoidsshowedemerging

Prox1

+

cellclusters2daysafter4-OH-Tamtreatment(Fig-

ureS1F).Atlatertimepoints,Prox1expressionwasmaintained

inisolatedcellclusters(FigureS1G).

ArecentstudyreportedthatPROX1ispartoftheWnt

high

CRC

stemcellgenesignature(deSousaEMeloetal.,2011).Wefound

thatProx1wasexpressedintheearliesthistologicaladenoma-

touslesions,theaberrantcryptfoci,oftheApc

min/+

mice,which

representawidelyusedmouseintestinaladenomamodel(Fig-

ure1A).ToexamineProx1expressionintheLgr5

+

intestinal

stemcellsoftheadenomas,weproducedApc

flox/flox

;Lgr5-

EGFP-IRES-Cre

ER

(LApc)miceandinducedApcdeletioninthe

stemcells(LApcD/D).LApcandLgr5-EGFP-IRES-Cre

ER

mouse

intestinesshowgreenEGFPsignalsintheLgr5

+

stemcellsatthe

cryptbottom(Barkeretal.,2007).Notably,thecyclinD1

high

cell

clusterswereenrichedmostlyamongtheLgr5

low

andLgr5

C0

cell

populationsintheintestinaladenomasafterApcdeletion,con-

firmingthatcyclinD1

high

cellsdonotaccumulateinthestem

cellpopulation(Figure1B).Prox1wasexpressedinsomeof

thePanethcellsintheileumandProx1

+

cellsinthenormalintes-

tinewereoftenlocatedneartheLgr5

+

cells(FigureS1H;Fig-

ure1C),buttherewasnooverlapbetweentheEGFPand

Prox1signalsintheuntreatedintestineorduringepithelialrepair

at4,6,and10daysafter6Gyirradiation(FigureS1Ianddatanot

shown).However,5daysaftertamoxifeninjection,Prox1

expressionwasobservedalsointheLgr5

+

cellsofthedevel-

opingadenomas(Figure1C).Takentogether,theseresults

indicatethatProx1isinducedduringtheearlystepsoftumori-

genesisinadenomacells,includingLgr5

+

stemcells,bothinvivo

andexvivo,butnotinthecyclinD1

high

cellsthatmayrepresent

moredifferentiatedorintestinalprogenitor-likecells.

ASubpopulationoftheProx1

+

CellsHasStemCell

ActivityinIntestinalAdenomas,butNotintheNormal

Intestine

TostudyifProx1

+

cellshavestemcellactivityinintestinalade-

nomas,weproducedProx1-Cre

ER

(Srinivasanetal.,2007);

Rosa26-tdTomato

flox/Stop/flox

;Apc

min/+

miceforlineage-tracing

experiments.Inthismodel,asingletamoxifeninjectionactivated

theCreallele,resultinginexpressionofthetdTomatoredfluo-

rescentproteinonlyintheProx1

+

tumorcells1dayafterthe

tamoxifeninjection(Figure1D),confirmingthattheactivation

oftheCreproteinisspecifictotheProx1-expressingtumorcells.

However,28daysaftertamoxifeninjection,weobservedthatthe

Prox1

+

cellshadproducedadjacenttdTomato

+

/Prox1

C0

prog-

eny,whichoccasionallystainedformucin2ofgobletcellsor

forlysozymeofPanethcells,indicatingthatProx1

+

tumorcells

cangiverisetodifferentiatedcellsintheintestinaladenomas

(Figures1Dand1E).

IntheProx1-Cre

ER

;Rosa26-tdTomato

flox/Stop/flox

;Apc

min/+

mice,Prox1

+

epithelialcellswereveryrarelylabeledoutsideof

thetumorsaftertamoxifeninjection.Inordertoactivatethe

lineagemarkermoreeffectively,weusedmiceharboringa

Prox1-Cre

ER

bacterialartificialchromosomeatanectopic

genomicsite(Bazigouetal.,2011).Inthesemice,onlysporadic

Prox1

+

intestinalepithelialcellswerepositivefortheredlineage

marker7daysaftertamoxifentreatment(Figure1F).Further-

more,Prox1deletionintheintestinalepitheliumofProx1

flox/flox

;

villin-Cre

ER

mice(VPD/D)didnotresultinanyobviousphenotype

(seeSupplementalResults).Thesedataindicatethatasubpop-

ulationoftheProx1

+

cellshasstemcellpropertiesinadenomas,

butnotinnormalintestine.

Prox1DeletionLeadstoLossofLgr5

+

StemCellsin

IntestinalAdenomas

ToanalyzethesignificanceofProx1specificallyinadenoma

stemcells,wedeletedApcandProx1intheLgr5

+

cellsof

Apc

flox/flox

;Prox1

flox/flox

;Lgr5-EGFP-IRES-Cre

ER

(LApcP)mice.

Inordertoobtainanefficientdeletion,tamoxifenwasinjected

during2consecutivedaysandthesizeoftheLgr5

+

cellpopula-

tionwasanalyzed21daysthereafter.Interestingly,themajority

oftheadenomascontainedsomeProx1

+

cells,suggestingan

incompletedeletionofProx1inthecryptstemcells(Figure2A).

However,theProx1

+

adenomascontainedfewerLgr5

+

cellsin

tamoxifen-treatedLApcP(LApcPD/D)mouseintestinesthanin

theLApcD/Dcontrols(Figure2B).Furthermore,thenumberof

stemcellswasevenlowerintheProx1

C0

crypt-likestructures

thatcontaineddislocatedLgr5

+

cellsbothinthesmallandlarge

intestine,indicatingthatsuccessfulProx1deletioninhibitsthe

expansionofLgr5

+

cellsintheadenomas(Figure2B).

Tofurthertestthishypothesisexvivo,weusedLApcD/Dand

LApcPD/Dorganoids.TheWnt-agonistR-Spondin1isrequired

forthesurvivalandgrowthoftheWT(LApcorLApcP)intestinal

organoids(Satoetal.,2009).WithoutR-Spondin1,onlyorgano-

idswithApcdeletionandresultingactiveb-catenin/TCF

pathwaysurvivebeyond4days(Wieneretal.,2014).Almostall

theProx1

+

cellsofLApcD/DorganoidswereLgr5-EGFPpositive

8daysaftertheadditionof4-OH-TamwithoutR-Spondin1(Fig-

ure2C).However,therewasamarkedreductioninthenumberof

viableorganoids8daysafterthesimultaneousdeletionofApc

1944CellReports8,1943–1956,September25,2014a2014TheAuthors

andProx1intheLgr5

+

cells(Figures2D–2F).Theviableorgano-

idscontainedsomeProx1

+

cellsatthistimepoint(Figure2C),

suggestingthatProx1deletionwasnotcompleteintheorgano-

ids.SinceProx1wasnotexpressedintheLgr5

+

stemcells

beforeApcwasdeleted,butonlyafter5daysofApcdeletion

invivo,thesedataindicatethatlossofProx1expressiondoes

Figure1.Prox1ExpressionandCellLine-

ageTracinginIntestinalAdenomasafter

ApcDeletion

(A)Prox1expressioninanaberrantcryptfocusin

theApc

min/+

intestine.

(B)Lgr5-EGFPandcyclinD1staininginLApc

mouseintestinebefore(LApc)and21daysafter

theadditionoftamoxifen(LApcD/D).Notesome

overlapofstainingintheLgr5

low

cellpopulation

(dottedareas).

(C)DistributionoftheLgr5-EGFPandProx1sig-

nalsintheintestinalepitheliumofLApcmiceafter

theinjectionofasingledoseoftamoxifen.Note

thelackofoverlapbetweenProx1

+

(arrowhead)

andLgr5

+

cellsatday0andtheoverlap(dotted

areas)atdays5and21.

(DandE)Immunostainingofadenomasderived

fromProx1-Cre

ER

;Rosa26-tdTomato

flox/Stop/flox

;

Apc

min/+

micefortheindicatedproteinsaftera

singletamoxifeninjection(celllineagetracing).

ThearrowsmarktdTomato

+

/Prox1

C0

/Mucin2

+

or

tdTomato

+

/Prox1

C0

/Lysozyme

+

cells.

(F)Prox1andtdTomatosignalsinProx1-Cre

ER

;

Rosa26-tdTomato

flox/Stop/flox

mice7daysafter

tamoxifeninjection.Thearrowsindicatetherare

labeledProx1

+

cellsinthenormalintestinal

epithelium.

Scalebarsrepresent20mm(BandD–F)or50mm

(AandC).SeealsoFigureS1.

notinfluencethetumorinitiationfre-

quency,butinstead,itinhibitstheex-

pansionoftheLgr5

+

adenomacell

population.

Todirectlyaddresstheconnectionbe-

tweenProx1andstemcellnumberin

anotherexvivoorganoidculturesystem,

weisolatedorganoidsfromApc

flox/flox

;

villin-Cre

ER

(VApc)andApc

flox/flox

;

Prox1

flox/flox

;villin-Cre

ER

(VApcP)mice,

whereApcand/orProx1aredeletedin

thewholeintestinalepithelium,including

theprogenitorcells.DeletionofProx1

alonefromthewholeintestinalepithelium

derivedfromtheProx1

flox/flox

;villin-Cre

ER

micedidnotinducemorphological

changesintheorganoids(FigureS2A).

However,deletionofbothApcand

Prox1(VApcPD/D)fromtheorganoids

increasedtherelativeexpressionlevelof

theprogenitormarkersNol1andWdr3

(VanderFlieretal.,2007)andtherelative

numberofcyclinD1

high

cellsincompari-

sonwithorganoidswithonlyApcdeletion

(Figures3A–3C).Asinthetamoxifen-injectedVApcD/Dmice,the

Prox1

+

cellswerecyclinD1

low

(Figure3B).Furthermore,RNAs

encodingtheWnt-targetintestinalstemcellsignaturegenes

Lgr5,Tnfrsf19,andAscl2weredecreaseduponProx1deletion,

whereasc-MycRNAwasnotchanged(Figure3Canddatanot

shown).MicroarrayanalysisofVApcD/DandVApcPD/Dcultures

CellReports8,1943–1956,September25,2014a2014TheAuthors1945

7daysaftertheadditionof4-OH-Tamindicateddecreasedin-

testinalstemcellmarkersandchangesintheexpressionlevels

ofspecificWnttargetsintheVApcPD/Dorganoids(FigureS2B).

ThesedatasuggestedthatProx1deletiondecreasesthenumber

ofadenomastemcells,resultinginaskewedenrichmentofthe

progenitorpopulationintheorganoids7daysafterApcdeletion.

Indeed,theProx1-deletedorganoidswerelessefficientinform-

ingneworganoidsubcultureswhentheyweredissociatedto

smallclusterscontainingapproximatelyfourtosevencells

(Figure3D).

PROX1SilencingDecreasesStemCellsinHumanCRC

Organoids

TomodeltheeffectofPROX1onstemcellactivityinCRCpro-

gression,wechosetousethehumanSW1222cellline(mutant

CRCgenesinthiscelllinearelistedinTableS1).Thiscellline

Figure2.Prox1DeletionfromtheLgr5

+

AdenomaCellsInhibitstheExpansionoftheStemCellPool

(A)Lgr5-EGFPandProx1signalsinuntreatedLApcmouseintestineandinLApcPD/Dintestine21daysaftertamoxifenaddition.ThearrowheadmarksaProx1

C0

cryptwithdislocatedLgr5

+

cells(implicatingApcdeletion).TheasterisksmarkProx1

+

crypts.

(B)ThenumberofLgr5

+

cellsperthevarioustypesofcryptsinthesmallintestineorcolon(Kruskal-Wallistest,n=25fromthreemice).Theboxplotindicatesthe

minimum,thefirstquartile,median,thirdquartile,andmaximum.

(C)Lgr5-EGFPandProx1signalsintheLApcD/DandLApcPD/Dorganoids.

(DandE)ViableLApcD/DandLApcPD/Dorganoids(D,arrowheads)andtheirquantification(E).

(F)Prox1RNAlevel.

For(C)–(F),thesampleswereanalyzed8daysaftertheadditionof4-OH-Tamand6daysaftertheremovalofthegrowthfactors.Scalebarsrepresent20mm(C)or

100mm(AandD).Mean+SDareshown(EandF).

1946CellReports8,1943–1956,September25,2014a2014TheAuthors

isenrichedforstemcellsthatcanself-renewanddifferentiate

intomultiplelineages(Yeungetal.,2010).Whilethe‘‘small

colonies’’inSW1222cell-derived3Dcultureshavealimited

growthpotentialandlacklumens,thelargeglandular‘‘megacol-

onies’’producecrypt-likestructuresconsistingofpolarizedcells

surroundingacentrallumen(Yeungetal.,2010).Lumensare

stemcell-dependentstructurespresentinwell-differentiated

tumorsandlikewiseinexvivoandinvitro3Dculturesfrom

humanandmouseadenomasandstemcell-containingCRC

celllines(Ashleyetal.,2013).

SincetheSW1222cellshaveheterogenousPROX1andnu-

clearb-CATENINexpressionlevels(FigureS2C),weisolated

subcloneswithalow(PROX1

low

)orahigh(PROX1

high

)percent-

ageofPROX1

+

cells(FigureS2D).WethensilencedPROX1in

thePROX1

high

clonebytwodifferentlentiviralshorthairpin

RNAs(shRNAs)(FigureS2Eanddatanotshown).Interestingly,

thePROX1-silencedSW1222-PROX1

high

cellsformedastrik-

inglyreducednumberofglandularcolonies(Figure3E),suggest-

ingthatPROX1silencinghasaprofoundeffectonCRCstemcell

activity.Furthermore,PROX1silencingdecreasedthenumber

oflumensintheglandularcoloniesderivedfromSW1222-

PROX1

high

cells(Figure3F).After12daysof3Dculture,the

LGR5andTNFRSF19RNAsweredecreased,whereasno

changeswereobservedintheexpressionofotherWNT-target

genes,suchasMYC(Figure3G).PROX1silencinginCRC

patient-derivedorganoidsalsoresultedindecreasedfrequency

ofnewlumen-containingorganoids(Figure3H).Notably,the

shPROX1organoidsformedsmallcolonieswithoutlumens(Fig-

ure3H).SimilarlytotheSW1222cellline,PROX1silencingin

CRCpatient-derivedorganoidsresultedinamarkedreduction

ofLGR5andTNFRSF19RNAlevels(Figure3I),eveninthe

presenceofaKRASmutation(seeSupplementalExperimental

Procedures).LGR5insituhybridizationfurtherindicatedthat

PROX1silencingdecreasesthenumberofCRCstemcells(Fig-

ures3Jand3K).Notably,therewasnodifferenceintheLGR5

RNAinPROX1-silencedSW1222cellsin2Dculture(datanot

shown),thusrulingoutthepossibilitythatLGR5isadirect

PROX1target.TheseresultssuggestthatlossofPROX1de-

creasestheCRCstemcellsalsoinhumanCRC,andthatthis

isindependentofthepresenceofKRASmutations.

PROX1

+

TumorCellsProliferateLessThanProx1

C0

TumorCells,yetPROX1DeletionLeadstoDecreased

CellProliferationintheTumorsInVivo

Totesttheinvivosignificanceofourfindings,weimplanted

SW1222-PROX1

high

cellsandtheirPROX1-silencedcounter-

partssubcutaneouslyintoimmunocompromised(NODscid

gamma[NSG])miceandmonitoredtumorgrowth.Similarly

totheProx1-deletedintestinaladenomas(Petrovaetal.,

2008),thePROX1-silencedtumorsgrewslowerthancontrols

transducedwithscrambledshRNA(Scr)atthisectopic

implantationsite(Figure4A).Notably,wedetected60%fewer

tumornestsinthePROX1-silencedsamplesascompared

toScrcontrols(200.4±46.8and70.9±11.8tumornests,

respectively;p<0.01)whentheywereexcised21daysafter

theimplantation.UnlikethescrambledshRNA-transduced

tumors,thePROX1-silencedtumorscontainedtumornests

withonlyfewglandularstructures(Figure4B).Similarlytothe

PROX1-silencedSW1222cells,subcutaneouslyimplanted

Prox1-deletedmouseVApcPD/Dorganoidsgrewslowerand

containedfewerglandsthanVApcD/Dorganoids(Figures4C

and4D).Prox1deletiondecreasedLgr5andTnfrsf19RNAs,

butnotofMycRNAinthetumors,confirmingourexvivore-

sults(Figure4E).

Intestinalepithelialprogenitorcellsproliferaterapidly,butonly

foralimitednumberofcellcycles(GregorieffandClevers,2005).

CarefulanalysisoftheProx1

+

andProx1

C0

cellsincontrol

SW1222-PROX1

high

orinVApcD/Dorganoid-derivedtumors

indicatedthattheKi67

+

proliferatingcellswereenrichedinthe

Prox1

C0

cellpopulation(FiguresS3AandS3B),suggestingthat

mostoftheProx1

+

cellsarenonproliferativeorslowlyprolifer-

ating.Interestingly,theProx1

+

/Lgr5

+

cellpopulationhadahigher

frequencyofKi67

+

cellsthantheProx1

+

/Lgr5

C0

populationinthe

LApcD/Dintestine(Figure4F),indicatingthattherapidlyprolifer-

atingProx1

+

cellsareenrichedintheLgr5

+

population.Similarly,

bromodeoxyuridinelabelingoftwohighlyproliferatingorganoid

cultures(VApcD/DandVCKID/D)showedthatonlyasmall

proportionoftheProx1

+

cellsproliferated(FigureS3C).VCKID/D

organoidsaredeletedoftp53andCsnk1a1,encodingcasein

kinaseIa(CKIa),whichphosphorylatesb-catenin,targetingit

toubiquitin-mediateddestruction;thus,theVCKID/Dorganoids

displayactivatedWntsignalingpathway(Elyadaetal.,2011).

WhereasProx1

+

cellsappearedtoproliferateslowly,we

observedadecreasedoverallfrequencyofKi67

+

tumorcellsin

subcutaneouslygrowingPROX1-silencedSW1222-PROX1

high

tumors,inProx1-deletedVApcPD/DorganoidtumorsinNSG

mice,andintheLgr5

+

cellsofLApcPD/Dintestinaltumors(Fig-

ure4G;FigureS3D).Apossibleexplanationfortheseapparently

contradictorydataisthatProx1deletionresultsindecreased

proliferationinthestemcellpopulation,whichleadstoexhaus-

tionoftheCRCstemcellpoolandconsequentlytoadecreaseof

theoverallratioofproliferatingcells.Indeed,aslowerprolifera-

tionrateofthePROX1-silencedSW1222-PROX1

high

cell-derived

organoidswasobservedonlyaftera14-dayculturingperiod,

whenthePROX1

+

organoidsalreadyhadanextensivelumen

formingactivity,butnotat6days(FigureS3E).Thisindicatesa

delayedeffectofPROX1silencingontheoverallcellproliferation

intheorganoids,inlinewiththeideathatPROX1influences

tumorgrowthbyregulatingthesizeoftheproliferatingstem

cellpool.

ThePROX1

-

(adherent)cellsoftheSW480CRCcellline

(SW480A)areunabletoinitiatesubcutaneoustumorsinNSG

mice,whereasthePROX1

+

cells(SW480R)areround,form

cellclusters,andaretumorigenic(Petrovaetal.,2008).To

testifPROX1silencinginalreadyestablishedCRCxenografts

regulatesstemcells,weimplantedcellsfromthePROX1

+

stemcell-likeSW480Rsubclone,expressingadoxycycline-

induciblePROX1shRNAconstruct(SW480R-sh)(Petrova

etal.,2008),intoNSGmice.Doxycyclinetreatmentwas

started8daysaftersubcutaneousinjection,whenthetumors

werealreadyvisible.Weobservedreducedgrowthofthe

PROX1-silencedtumorsafterday16(Figures4Hand4I),ata

timepointwhentheLGR5RNAlevelhadalreadymarkedly

decreasedinthedoxycycline-treatedtumors(Figure4J).These

datasuggestthatPROX1regulatesstemcellsalsoinestab-

lishedtumors.

CellReports8,1943–1956,September25,2014a2014TheAuthors1947

Figure3.Prox1DeletionLeadstoReducedStemCellActivityinExVivoOrganoids

(A)Theschematicoutlineofthemouseorganoidexperiments.GF,growthfactors.

(B)Prox1andcyclinD1immunostainingofsectionsfromVApcD/DandVApcPD/Dorganoids7daysafterthedeletionand5daysafterremovingR-Spondin1from

theculturemedium.

(C)RNAlevelsofprogenitormarkers(Nol1,cyclinD1,andWdr3),stemcellmarkers(Lgr5andTnfrsf19),andMycinVApcD/DandVApcPD/Dorganoidsanalyzed

byreal-timequantitativePCR(qRT-PCR).

(D)TheorganoidinitiatingfrequencyofVApcD/DandVApcPD/Dorganoids.

(E)Theproportionofglandularcoloniesderivedfrom1,000SW1222-PROX1

high

cells(transducedwithScr,sh1,orsh2PROX1shRNAlentivirus).

(F)ThenumberoflumensintheglandularcoloniesderivedfromScrorshPROX1-transduced(sh1,sh2)SW1222-PROX1

high

cellsatday14(n=10forsh1and

n=12forsh2).Thelumens(asterisks)weredetectedbyphalloidinstainingandcountedinopticalsectionsofconfocalmicroscopicimages.Theboxplotindicates

theminimum,thefirstquartile,median,thirdquartile,andmaximum.

(G)Real-timeqPCRoftheindicatedRNAsfromSW1222-PROX1

high

cell-derivedorganoids,transducedwithshPROX1orScrlentivirusandgrowninMatrigelfor

14days.

(legendcontinuedonnextpage)

1948CellReports8,1943–1956,September25,2014a2014TheAuthors

PROX1SilencingIncreasesAnnexinA1Expressionin

MultipleCRCModels

Todeterminewhichgenesareresponsiblefortheeffectof

PROX1ontheadenoma/CRCstemcells,wetestedPROX1-

(H)Imagesandtherelativelumen-containingorganoidinitiatingfrequencyofScrandshPROX1lentivirus-transducedhumanorganoidsderivedfromCRC

patients.TheGFPsignalshowsthelentiviraltransductionefficiency.

(I)RNAlevelsoftheindicatedgenesinhumanCRCpatient-derivedorganoids,transducedwithScrorshPROX1lentivirus.Notethatwhilesample1containsthe

G12DclinicallyrelevantmutationintheKRASgene,samples2and3arewild-type.

(JandK)LGR5insituhybridizationandquantificationofLGR5

+

cellsintheScrandshPROX1-transducedSW1222-PROX1

high

clone.

Scalebarsrepresent100mm(B)or50mm(E,F,H,andK).Mean+SD(C–EandG–I)ormean+SEM(J)areshown.SeealsoFigureS2.

Figure4.PROX1RegulatestheNumberof

StemCellsviaCellProliferation

(A)Growthcurveofsubcutaneoustumorsderived

fromScrorshPROX1-transducedSW1222-

PROX1

high

cellsinNSGmice(n=10).

(B)PROX1andE-CADHERINimmunostainingand

HEstainingoftumorsections21daysaftersub-

cutaneousinjectionofSW1222-PROX1

high

cells

intoNSGmice.Thedashedlineindicatesthetu-

morborder,thearrowheadspointtodegenerating

glandularstructuresandtheasteriskmarks

necroticarea.

(C–E)Immunostaining,tumorvolume(CandD)

andRNAquantification(E)ofVApcD/D(black

columns)andVApcPD/D(redcolumns)organoid-

derivedtumors12and28daysaftertheirsubcu-

taneousinjectionintoNSGmice(confocal3D

reconstruction).Notethemorecomplexglandular

structureoftheVApcD/Dtumors(asterisks).

(FandG)Thepercentageofproliferating(Ki67

+

)

cellsamongtheProx1

+

cellsintheLgr5

+

and

Lgr5

C0

populationsintheintestinalepitheliumof

LApcD/Dmice21daysafterApcdeletion(n=10–

12)(F).ThepercentageofKi67

+

cellsintheLgr5

+

cellpopulationintheintestinalepitheliumof

LApcD/DandLApcPD/Dmice(n=9–11)(G).The

boxplotsindicatetheminimum,thefirstquartile,

median,thirdquartile,andmaximum(FandG).

(H–J)GrowthofsubcutaneousSW480R-shtu-

morsinNSGmice(H)withorwithoutdoxycycline

(n=10).Doxycylinetreatmentfromday8(blue

arrow)today16(redarrow)aftercellimplantation

wasusedforthesilencingofPROX1.Immuno-

staining(I)andquantitativeRT-PCR(qRT-PCR)(J)

analysesatday16.

Scalebarsrepresent50mm(B)and100mm(B,D,

andI).Mean+SD(C,E,andJ)ormean+SEM

(AandH)areshown.SeealsoFigureS3.

regulatedcandidategenesbasedon

themicroarraydataderivedfromthe

SW480Rsubclone(Petrovaetal.,2008).

Basedonourinitialresults,wefocused

furtheronthecalcium-dependentphos-

pholipidbindingproteinAnnexinA1

(ANXA1),whichhasbeenshowntoinhibit

breastcancermetastasis(Maschleretal.,

2010).PROX1suppressedANXA1RNA

(4.21±0.08-fold,mean±SD)andprotein

expressioninSW480R-shcells(Fig-

ureS4A).Anxa1wasincreasedafter4-OH-Tamadditionto

VApcorganoidsandevenfurtherelevatedwhenalsoProx1

wasdeleted(Figure5A;FiguresS4BandS4C),suggestingthat

Prox1suppressesAnxa1expression.Furthermore,theProx1

C0

CellReports8,1943–1956,September25,2014a2014TheAuthors1949

(legendonnextpage)

1950CellReports8,1943–1956,September25,2014a2014TheAuthors

cellsshowedmoreintenseAnxa1stainingthanProx1

+

cellsin

tumorsectionsfromApc

min/+

mice(FigureS4D).

WeobservedamutuallyexclusivestainingforAnxa1and

Prox1intumorsfromVCKID/DmiceandinhumanCRCsamples

(Figures5Band5C).Furthermore,theLgr5

+

intestinalstemcells

intheLApcmiceshowedverylittleornoepithelialAnxa1expres-

sionbeforeApcdeletion(FigureS4E).Theycontainedalowlevel

ofAnxa1aftertamoxifeninjection,whereastheAnxa1

high

cells

werenegativeforLgr5intheresultingadenomas(FigureS4F).

Importantly,transfectionofadominant-negativeTCF4(tran-

scriptionfactor7-like2[TCF7L2])construct,aWntpathwaysup-

pressortothePROX1

C0

subcloneoftheSW480CRCcellline

(SW480A)(Petrovaetal.,2008),ledtosuppressionofANXA1

expression(Figure5D).ThesedataindicatethatAnxa1ismini-

mallyexpressedinthenormalintestinalepithelium,wherethe

activationoftheWntpathwayafterApcdeletionincreases

Anxa1expression,whichisthensuppressedbytheinduction

ofProx1inthetumorcells.

ANXA1SilencingMimicstheEffectsofPROX1inCRC

WenexttestedthepossibilitythatthestrongANXA1suppression

mediatestheeffectsofPROX1,suchastheassociatedrear-

rangementoftheactincytoskeleton,changesincellshape,

andadherencetothecultureplates(Petrovaetal.,2008).Indeed,

ANXA1overexpressionintheSW480Rsubcloneinducedan

elongatedcellshapeandincreasedthenumberoftightly

adherentcells,whereasANXA1suppressionresultedinrear-

rangementoftheactincytoskeleton,roundedcellshape,and

decreasedadherence(FiguresS5A–S5C).Interestingly,Anxa1

silencinginVApcD/DorganoidsincreasedtheLgr5andTnfrsf19

stemcellmarkerRNAs,withoutaffectingMyc(Figure5E).

AlthoughANXA1silencinghadnoeffectonPROX1expression

inSW1222-PROX1

low

orSW480cellsin2Dculture(datanot

shown),itresultedinincreasedlumenformation(Figure5F),

upregulationofTNFRSF19andLGR5RNAs,andincreaseof

LGR5

+

cellsin3DorganoidsderivedfromshANXA1-transduced

cells(Figures5Gand5H).Incontrast,ANXA1overexpression

inSW1222-PROX1

high

cellsdecreasedtheproportionofglan-

dularcolonies,thenumberoforganoidlumens,andLGR5

andTNFRSF19RNAlevelswhencomparedtothecontrols(Fig-

uresS5D–S5F).Furthermore,theANXA1-silencedSW1222-

PROX1

low

cell-derivedtumorsgrewfasterthancontroltumors,

containedmoreKi67

+

cells,andwerecomposedoflarger

tumornestswithmultiplelumensorganizedintolabyrinth-like

structures(Figure5I;FigureS5G).Similartotheeffectof

PROX1,ANXA1silencinginSW1222cell-derivedorganoids

wasassociatedwithenhancedcellproliferationonlyafter

16days,whentheorganoidsshowedextensiveoutpocketing

andlumenformation,butnotatday6(FigureS5H).Thesedata

indicatethatANXA1silencingmimicstheeffectsofPROX1

expression,leadingtoanexpansionofthestemcellpoolvia

increasedproliferation.

SilencingtheActin-BindingProteinFILAMINA

DecreasesStemCellsinCRC

ReanalysisofthemicroarraydataderivedfromtheSW480R

subcloneshowedthatfocaladhesionandregulationofactin

cytoskeletonwerethetopKyotoEncyclopediaofGenesand

GenomesgenepathwaycategoriesaffectedwhenPROX1was

silenced(datanotshown).Thus,wesearchedforPROX1-regu-

latedgenesdirectlyaffectingthecytoskeletonamongpublished

CRCstemcellgenesets(deSousaEMeloetal.,2011)andprog-

nosismarkersinCRCsubgroups(Sadanandametal.,2013).We

foundthegeneencodingtheactin-bindingproteinfilaminA

(FLNA)inbothgenesets.FLNAconnectsactinfilamentsto

transmembranereceptors,includingb1integrin,modulatescell

migration,andfunctionsasacentralmechanotransduction

elementofthecytoskeleton(Ehrlicheretal.,2011;Zhouetal.,

2010).Interestingly,wefoundhighlyincreasedFlnaexpression

intheProx1

+

andLgr5

+

tumorcells(Figure6Aanddatanot

shown)andacorrelationbetweenPROX1andFLNAlevelsin

theSW1222-PROX1

high

cell-derivedorganoids(FigureS5I).

FLNAwasalsoupregulatedintheANXA1-silencedorganoids

(Figures6Band6C).Furthermore,PROX1silencinginthe

SW1222-PROX1

high

andCRCpatient-derivedorganoidsre-

sultedinmarkedlydecreasedFLNARNA(Figure6D)andprotein

(Figure6E).

BecausebothPROX1andANXA1regulatedFLNAexpression

inintestinaladenomasandinCRC3Dorganoids,westudied

FLNAfunctionbysilencingitsexpressioninVApcD/Dorganoids

andinSW1222-PROX1

high

cells.ThisledtoreducedLgr5and

Tnfrsf19RNAs(Figure6F),toadecreaseofSW1222cell-

derivedglandularcoloniesandlumens(Figure6G),andtoa

decreasednumberofKi67

+

cellsintheorganoids(Figure6H).

Surprisingly,FLNAsilencingdidnotaffectthegrowthofthecells

in2Dcultureconditions(datanotshown).Overall,theseresults

suggestamodelwherethelossofProx1leadstoanelevated

Anxa1levelandareductioninFlna,whichlimittheexpansion

Figure5.ANXA1SilencinginCRCLeadstoElevatedStemCellActivityandTumorGrowth

(A)ImmunostainingofVApcD/DandVApcPD/Dorganoidsfortheindicatedproteins.

(BandC)Immunoperoxidasestainingofsectionsfrom(B)VCKID/Dtumorsand(C)fromatumorofaCRCpatient(n=5).

(D)ANXA1expressioninSW480cellstransfectedwithadominant-negativednTCF4-FLAGoranunrelatedcontrolconstruct(TIE1-FLAG).Thearrowheads

indicatetransfectedcells.

(E)RNAanalysisofScrandshAnxa1-transducedVApcD/Dorganoids(blackandgreencolumns,respectively).

(F)NumberoflumensinSW1222-PROX1

low

subclone-derivedorganoids,transducedwitheitherScrorshANXA1-expressinglentivirus,stainedwithfluorescent

phalloidinandcountedfromopticalsectionsofconfocalimages.Thewhiteasterisksmarkthelumens(n=11forsh(1)andn=25forsh(2)).Theboxplotindicates

theminimum,thefirstquartile,median,thirdquartileandmaximum.

(GandH)RNAlevelsoftheindicatedtranscripts(G)andLGR5insituhybridization(H)(redsignal)inSW1222-PROX1

low

-derivedorganoids,growninMatrigel

for16days.

(I)Growthcurves(n=6),histologicalanalysisandimmunostainingofSW1222-PROX1

low

tumorsgrowingsubcutaneouslyinNSGmice.

Forthestaining,thetumorswereisolatedonday11aftertheimplantation.Scalebarsrepresent100mm(A–D)or50mm(F,H,andI).Mean+SD(EandG)or

mean+SEM(HandI)areshown.SeealsoFiguresS4andS5.

CellReports8,1943–1956,September25,2014a2014TheAuthors1951

oftheadenoma/CRCstemcellpopulationviadecreasingcell

proliferation.

PROX1SilencingReducesCellSurvivalinHypoxic

TumorXenografts

Interestingly,whengrowntoalargesizeinMatrigelculture

(>150mmdiameter,>5daysaftersubculture)orinjectedsubcu-

taneouslyintoNSGmice,Prox1-deletedVApcPD/Dorganoids

containedmoreapoptoticcellsthanVApcD/Dorganoids(Fig-

ure7A).Also,wedetectedanintensestainingfortheapoptosis

markeractiveCASPASE-3insidethePROX1-silencedSW1222-

PROX1

high

cell-derivedsubcutaneouslygrowingtumorsthat

werepositiveforthehypoxiamarkercarbonicanhydraseIX,

butnotatthetumormargin(Figure7Banddatanotshown),sug-

gestingreducedtumorcellsurvivalintheischemic/hypoxic

tumorinterior.Interestingly,wefoundnodifferenceintheendo-

mucin

+

bloodvesseldensitybetweenScrandshPROX1tumors,

evenwhenvascularendothelialgrowthfactor(VEGF)wasover-

expressedinthetumorcells(FiguresS6AandS6B).However,

theshPROX1tumorsweresmallerthantheScrtumors,

evenwhenVEGFwasoverexpressedinthetumorcellsand

VEGFfailedtorescuethelargenecroticareasinsidethe

Figure6.SilencingtheActin-BindingProteinFilaminAInhibitsStemCellActivity

(A)ImmunostainingforfilaminA(Flna)andProx1inanintestinaladenomaofApc

min/+

mice.

(BandC)FLNAstaining(B)andqRT-PCR(C)fromscrambledandshANXA1-transducedSW1222-PROX1

low

cell-derivedorganoids14daysafter3Dseeding.

(D)FLNARNAinCRCpatient-derivedandSW1222-PROX1

high

cell-derivedorganoids,transducedwithScrorshPROX1lentivirus(qRT-PCR).

(E)ImmunostainingforPROX1andFLNAinScrorshPROX1-transduced,SW1222-PROX1

high

cell-derivedorganoids,culturedfor7daysinMatrigel(confocal3D

reconstructions).

(F)ThelevelsoftheindicatedRNAsinVApcD/Dorganoids10daysaftertransductionwithScrorshFlnalentivirus(qRT-PCR).

(G)Theproportionofglandularcoloniesandthenumberoflumens(n=8forsh(1)andn=14forsh(2))inSW1222cell-derivedorganoids,transducedwiththe

indicatedlentivirus.

(H)ThepercentageofKi67

+

cellsinScr(blackbox)orshFLNA-transduced(graybox)SW1222cell-derivedorganoidsafter9daysof3Dculturing,countedfrom

confocalopticalsections(n=12).

Theboxplotsin(G)and(H)indicatetheminimum,thefirstquartile,median,thirdquartile,andmaximum.Mean+SDareshown(C,D,F,andG).Scalebars

represent50mm(AandB)or25mm(E).SeealsoFigureS5.

1952CellReports8,1943–1956,September25,2014a2014TheAuthors

PROX1-silencedtumors(FiguresS6AandS6B).Thus,thepres-

enceofthelargenecroticareasintheshPROX1tumorscould

notbeexplainedbythelackofangiogenicfactors.

AlthoughANXA1silencingmimickedseveraloftheeffectsof

PROX1intheintestinaladenomaandCRCmodels,the

apoptosisrateintheANXA1-silencedsubcutaneouslygrowing

tumorswasnotsignificantlyaffectedwhenanalyzed9or

24daysafterimplantation(datanotshown).Thisraisedthepos-

sibilitythatinunfavorableconditionsalsoalternativepathways

contributetotheapoptosisintheshPROX1tumors.Autophagy

isanessentialcellularprocessforthesurvivalofcellsunderhyp-

oxiaornutrientdeprivation(Satoetal.,2007).Interestingly,

PROX1silencingintheSW480Rcellsresultedinadecreaseof

theautophagy-associatedLC3-IIproteinbothinnormalmedium

andinstarvationconditions(Figures7Cand7D).Furthermore,

lackofPROX1preventedtheaccumulationoftheLC3-contain-

ingearlyautophagosomesinthepresenceofthelysosomalin-

hibitorbafilomycinAorchloroquine,whichinhibitthefusionof

autophagosomeswiththelysosomes(Figure7D).Importantly,

weobservedadecreasednumberofLC3-containingearlyauto-

phagosomesunderhypoxiainlentivirallyPROX1-silenced

SW480RorSW1222-PROX1

high

cellswhentheirfusionwithlyso-

someswasinhibited(Figure7E),suggestingthatPROX1en-

hancestheautophagicflux.Theadditionofalowconcentration

ofchloroquineorbafilomycinAresultedinlossoftheLgr5

+

cells

afterApcdeletion(Figure7F).Ofnote,thischloroquineconcen-

trationinhibitedtheformationofVApcD/Dorganoidcolonies

uponsubculture,butnottheirsurvivalwhenadded2daysafter

organoidplating(Figure7G).Furthermore,theadditionofchloro-

quineorbafilomycinatalowdosetotheLApcD/Dorganoidcul-

turesreducedtheproportionofviableorganoidstothesame

levelasProx1deletionintheLApcPD/Dcultures,thushigh-

lightingtheimportantroleofautophagyinpromotingtumorcell

survivalspecificallyintheProx1

+

cells(Figure7H).

DISCUSSION

Prox1iscriticalforthefateofseveraltypesofstemandprogen-

itorcells(Elsiretal.,2012).Here,weshowthatProx1isnotex-

pressedintheLgr5

+

stemcellsofnormalintestinalcrypts,but

isinducedsoonaftertheinitiatingmutationinintestinaltumori-

genesis.WeshowthattheProx1

+

cellsgiverisetomorediffer-

entiatedProx1

C0

cellsintheadenomas,butnotinthenormal

intestine,indicatingthatasubpopulationoftheProx1

+

cells

hascancerstemcellactivity.UponProx1deletion,thenumber

ofstemcellsdeclinedandthiswasreflectedlateronas

decreasedoveralltumorcellproliferationandtumorgrowth.

Stem-likecellsprovideanimportantdrugtargetincancer,as

theyareabletopersistintumorsasadistinctpopulation,self-

renew,anddifferentiate,andtheyareassociatedwithtumor

relapse(SampieriandFodde,2012).Therefore,development

ofspecifictherapiestargetedatcancerstemcellsmayimprove

survival.Inelegantstudies,Schepersetal.demonstratedthat

theLgr5

+

cellsinintestinaladenomashavestemcellproperties

(Schepersetal.,2012).Interestingly,recentstudiesindicated

thatintestinaladenomascontainfewerstemcellsthanLgr5

+

cells,suggestingthatonlyasubpopulationoftheLgr5

+

cells

functionasstemcells(Kozaretal.,2013).Furthermore,Myant

etal.haveshownthattheRac1GTPaseisanimportantregulator

oftheproliferationoftheLgr5

+

cellpopulationinintestinalade-

nomas(Myantetal.,2013).AlthoughtheLgr5

+

cellsmaybe

dispensableforhomeostasisofthenormalintestineandfor

increasedproliferationofintestinalepitheliumafterApcdeletion

(Metcalfeetal.,2014),theexpansionofcellsderivedfromthe

Lgr5

+

cellsinadenomasafterApcdeletionclearlyindicates

thattheycontributetotumorgrowth(Schepersetal.,2012).

Importantly,Prox1deletionnotonlydecreasedthenumberof

Lgr5

+

cellsinintestinaladenomasinvivo,butitalsoreducedtheir

proliferation,theorganoid-initiatingfrequencyandthenumberof

megacoloniesandlumensintheglandularCRCorganoids,

whichallrepresentindicatorsofcancerstemcells(Ashley

etal.,2013).Furthermore,PROX1silencingafterthetumor

establishmentresultedinadecreaseoftheLGR5

+

tumorcell

markerbeforeadeclineintheoveralltumorgrowthrate.

Mechanistically,weshowthatthephospholipid-bindingpro-

teinAnxa1isincreasedbytheWntpathwayactivationafter

Apcdeletion,whileProx1suppressesitsexpression.ANXA1

andPROX1showedalsomutuallyexclusiveexpressionpatterns

inhumanCRCsamples.Strikingly,thesilencingofANXA1inthe

PROX1

C0

cellsmimickedtheeffectsofPROX1intheCRCcells.

Amongitsothereffects,ANXA1inhibitstheproinflammatory

phospholipaseA2,whichhasbeenshowntostimulatetheprolif-

erationofCRCcellsbyproducingvariouslipidmediatorsand

whichregulatesintercellularjunctionsandtheactincytoskeleton

(Cristanteetal.,2013;ParenteandSolito,2004;Surreletal.,

2009).BothANXA1silencingandPROX1expressionincreased

stemcellmarkersandthenumberofLGR5

+

cellsintumororga-

noidsandenhancedlumenformation,proliferationoftumorcells,

andtumorgrowthinvivo.Furthermore,ANXA1silencingand

PROX1expressionincreasedFLNA,whichstabilizesthecortical

3Dactinnetworks,linksthemtothetransmembranereceptorb1

integrin,andfunctionsasacentralmechanotransduction

elementofthecytoskeleton(Ehrlicheretal.,2011;Zhouetal.,

2010).Theroleoftheactincytoskeletonintheexpansionofthe

adenomastemcellpopulationwassupportedbyourFLNAre-

sults,showingthatsilencingthisactin-bindingprotein,which

wasexpressedintheLgr5

+

stemcellsofintestinaladenomas,

dramaticallyinhibitedtheactivityandproliferationoftheade-

noma/CRCstemcellsin3D,butnotin2D,cultureconditions.

FLNAisoneofthemarkersofaCRCsubtypecharacterizedby

poordisease-freesurvival(Sadanandametal.,2013).FLNA

expressionisincreasedinanumberofcancers,andithas

beenrecentlyshowntoboostthehypoxiaresponseandtumor

progression(Zhengetal.,2014).Consistentwiththis,the

FLNA-expressingPROX1

+

tumorcellsweremoreresistantto

apoptosisthantheFLNA

low

PROX1

C0

tumorcellsbothinvivo

andintheorganoids.Therewasastrikingincreaseinapoptosis,

particularlyinthecentralpartsofthePROX1-silencedtumors

growingsubcutaneouslyinmice.However,thesensitivityofthe

PROX1

C0

tumorcellstoapoptosiswasnotduetoinsufficient

expressionofangiogenicgrowthfactors,asshownbythe

inabilityofVEGFoverexpressiontorescuethedifference.

Instead,theanalysisofautophagymarkersanduseoflysosomal

inhibitorsofautophagyindicatedthatPROX1expressionsus-

tainsautophagy,whichisknowntobeessentialforthesurvival

ofCRCcells(Satoetal.,2007).Thisfindingisparticularly

CellReports8,1943–1956,September25,2014a2014TheAuthors1953

Figure7.LossofPROX1ReducesCRCStemCellSurvivalunderUnfavorableConditions

(A)Percentageofcaspase-3

+

apoptoticcellsamongtheE-cadherin

+

epithelialcellsinVApcD/D(blackcolumns)andProx1-deleted(VApcPD/D;redcolumns)

organoids8daysaftertheadditionof4-OH-Tamand6daysaftertheremovalofR-Spondin1(rightpanel,n=14)orgrowingsubcutaneouslyinNSGmice

(leftpanel,n=22).

(B)Activecaspase-3stainingofsubcutaneouslygrowingScrandshPROX1-transducedSW1222-PROX1

high

tumorsinNSGmice.NotethattheshPROX1-

transducedtumorinteriorishighlyapoptotic(asterisk).

(CandD)ImmunoblottingofSW480R-shcellsfortheindicatedproteins.Cellswereeitherculturedincompletemedium(C)orinstarvationmediumlackingamino

acids8hrbeforeproteinisolation(D)intheabsenceorpresenceof100nMbafilomycinAor30mMchloroquine.

(E)ThenumberofLC3

+

granulesintheindicatedcelllinestransducedwithScrorshPROX1lentivirusandculturedinstarvationmediumcontaining100nM

bafilomycininhypoxiafor8hr(n=50).

(F)FlowcytometricanalysisoftheLgr5-EGFP

+

cellsinLApcD/Dorganoids,9daysaftertheApcdeletionand3daysaftertheadditionof15mMchloroquineor

0.2mMbafilomycinA.

(G)TheplatingefficiencyandsurvivalpercentageofVApcD/DorganoidsinthepresenceorabsenceofchloroquineorbafilomycinA.Notethatfororganoid

survivalchloroquineorbafilomycinwereadded2daysafterorganoidsplittingfor3days.

(legendcontinuedonnextpage)

1954CellReports8,1943–1956,September25,2014a2014TheAuthors

interestingconsideringthathypoxia-inducedautophagycanpro-

motetumorcellsurvivalandadaptationtoantiangiogenictreat-

ment,whichisusedinCRCtherapy(Huetal.,2012).Ourresults

areinagreementwiththefindingsofRagusaetal.,whoshowthat

PROX1promotesthemetabolicadaptationofCRCcellsinunfa-

vorablemicroenvironments,andthuscriticallycontributestothe

metastaticoutgrowthofCRCs(Ragusaetal.,2014).

Insummary,weshowherethatPROX1isinducedinintestinal

stemcellsinadenoma/CRCsoonaftertheactivationoftheWnt

pathway.WhiletheAPCmutationinducesbothPROX1and

ANXA1expressionintheepithelium,PROX1restrictsANXA1

levelsandinducesFLNA,whichstimulatescellproliferation

andpromotesstemcellactivityintheadenomas,andmaycoun-

teractstemcellexhaustionduringtumorgrowth.Theneteffectis

theexpansionoftheadenoma/CRCstemcellpopulationand

increasedtumorgrowth.Inthehypoxicpartsoftumortrans-

plants,PROX1promotestumorcellsurvivalbyincreasingauto-

phagy.Basedonthisstudy,PROX1regulatesthenumberof

adenoma/CRCstemcellswithoutaffectingthehomeostasisof

thenormalintestine,thusprovidinganattractivetherapeutic

targetpathwayfordrugdevelopmentinCRC.

EXPERIMENTALPROCEDURES

AdetaileddescriptionoftheexperimentalproceduresisprovidedinSupple-

mentalExperimentalProcedures.

IntestinalCrypt/OrganoidCultures

TheNationalBoardforAnimalExperimentsattheProvincialStateOfficeof

SouthernFinlandapprovedallexperimentsperformedwithmice.Intestinal

cryptsfromApc

flox/flox

;villin-Cre

ER

,Apc

flox/flox

;Prox1

flox/flox

(Harveyetal.,

2005);villin-Cre

ER

,Csnk1a1

flox/flox

;tp53

flox/flox

;villin-Cre

ER

(Elyadaetal.,

2011),Apc

flox/flox

;Lgr5-EGFP-IRES-Cre

ER

(Barkeretal.,2007)andApc

flox/flox

;

Prox1

flox/flox

;Lgr5-EGFP-IRES-Cre

ER

micewereisolatedandculturedas

describedpreviously(Satoetal.,2009,2011).Toactivatetheendogenous

b-catenin/TCFpathwayinmouseorganoids,culturesweretreatedwith

300nM4-hydroxy-tamoxifen(4-OH-Tam)for48hr.Organoidswiththeendog-

enouslyactiveb-catenin/TCFpathwaywerethenselectedandculturedin

growthfactor-deficientmedium.

HumanOrganoidCultures

For3Dculture,SW1222orpatient-derivedCRCcellswereextensivelytrypsi-

nized,embeddedintoMatrigel(500–2,000cells/50mlMatrigel/well),and

grownfor3–16days.TheethicscommitteeoftheDepartmentofSurgeryat

HelsinkiUniversityHospitalapprovedallexperimentsinvolvingpatientsam-

ples,andinformedconsentwasobtainedfromthepatients.Tissuesamples

isolatedfromCRCpatientswereprocessedaccordingtoapreviouslypub-

lishedmethod(Satoetal.,2011).

InVivoExperiments

Micewereinjectedwith2mgtamoxifen(Sigma)dissolvedin200mlsunflower

oil(Sigma)attheageof8–9weeks.Themicewereeuthanizedattheindicated

timepoints.ThemicewereontheC57Bl/6background.Inallexperiments

littermatecontrolswereused.

StatisticalAnalysis

Statisticalcomparisonoftwogroupswasdonebytwo-tailedunpairedor

pairedttestusingtheSPSSsoftwareunlessotherwiseindicated.Fornonpara-

metrictests,theMann-WhitneyUtestwasused,andthedataarepresentedas

boxplots,showingthefivestatistics(minimum,firstquartile,median,third

quartile,maximum).Thestatisticalsignificanceismarkedbyp<0.05,p<

0.01,andp<0.005.

ACCESSIONNUMBERS

TheGeneExpressionOmnibusaccessionnumberforthemicroarraydatare-

portedinthispaperisGSE47568.

SUPPLEMENTALINFORMATION

SupplementalInformationincludesSupplementalResults,Supplemental

ExperimentalProcedures,sixfigures,andonetableandcanbefoundwith

thisarticleonlineathttp://dx.doi.org/10.1016/j.celrep.2014.08.034.

AUTHORCONTRIBUTIONS

Z.W.,J.H.,V.H.,A.M.B.andK.A.designedresearch;Z.W.,J.H.,V.H.,A.M.B.,

P.K.,T.H.,andO.D.performedresearch;C.H.,O.K.,G.O.,andY.B.-N.contrib-

utednewreagents/analytictools;Z.W.,J.H.,V.H.,andK.A.analyzeddata;and

Z.W.,J.H.,andK.A.wrotethepaper.

ACKNOWLEDGMENTS

WethankDr.LeifAnderssonfortheconsultationsonhistopathology,Dr.Taija

Ma¨kinenfortheProx1-Cre

ER

mice,Dr.DarrenTysonfortheANXA1-modu-

latingconstructs,Dr.DavidCalderwoodfortheFLNAantibody,Dr.Meenhard

Herlyn(WistarInstitute)fortheSW1222cellline,Dr.TatianaPetrovafordiscus-

sionsofPROX1functionsinCRC,Dr.PekkaKatajisto,Dr.TuomasTammela,

andDr.TimoOtonkoskiforcommentsonthemanuscript,andLariPyo¨ria¨,Kirsi

Lintula,KatjaSalo,LauraRaitanen,andTapioTainolafortheirhelpwiththeex-

periments.TheBiomedicumImagingUnitisacknowledgedformicroscopy

services.ThisworkwasfundedbytheSigridJuseliusFoundation,theFinnish

CancerOrganizations,andtheAcademyofFinland(262976).Z.W.wassup-

portedbytheSigridJuseliusFoundationandbytheMarie-CurieIntra-Euro-

peanFellowship(PIEF-GA-2009-236695).

Received:October23,2013

Revised:January14,2014

Accepted:August17,2014

Published:September18,2014

REFERENCES

Ashley,N.,Yeung,T.M.,andBodmer,W.F.(2013).Stemcelldifferentiationand

lumenformationincolorectalcancercelllinesandprimarytumors.Cancer

Res.73,5798–5809.

Barker,N.,vanEs,J.H.,Kuipers,J.,Kujala,P.,vandenBorn,M.,Cozijnsen,

M.,Haegebarth,A.,Korving,J.,Begthel,H.,Peters,P.J.,andClevers,H.

(2007).Identificationofstemcellsinsmallintestineandcolonbymarker

geneLgr5.Nature449,1003–1007.

Barker,N.,Ridgway,R.A.,vanEs,J.H.,vandeWetering,M.,Begthel,H.,van

denBorn,M.,Danenberg,E.,Clarke,A.R.,Sansom,O.J.,andClevers,H.

(2009).Cryptstemcellsasthecells-of-originofintestinalcancer.Nature

457,608–611.

Batlle,E.,Henderson,J.T.,Beghtel,H.,vandenBorn,M.M.,Sancho,E.,Huls,

G.,Meeldijk,J.,Robertson,J.,vandeWetering,M.,Pawson,T.,andClevers,

H.(2002).Beta-cateninandTCFmediatecellpositioningintheintestinal

epitheliumbycontrollingtheexpressionofEphB/ephrinB.Cell111,251–263.

(H)RelativesurvivalrateofLApcD/DorLApcPD/Dorganoids8daysafter4-OH-Tamand6daysinR-Spondin1-freemedium.Chloroquine(15mM)orbafilomycin

A(0.2mM)wereadded4daysafter4-OH-Tamfor4days.

Theboxplotsin(A)and(E)indicatetheminimum,thefirstquartile,median,thirdquartile,andmaximum.Mean+SDareshownfor(F)–(H).Scalebarsrepresent

100mm.SeealsoFigureS6.

CellReports8,1943–1956,September25,2014a2014TheAuthors1955

Bazigou,E.,Lyons,O.T.,Smith,A.,Venn,G.E.,Cope,C.,Brown,N.A.,and

Makinen,T.(2011).Genesregulatinglymphangiogenesiscontrolvenousvalve

formationandmaintenanceinmice.J.Clin.Invest.121,2984–2992.

Clevers,H.(2006).Wnt/beta-cateninsignalingindevelopmentanddisease.

Cell127,469–480.

Cristante,E.,McArthur,S.,Mauro,C.,Maggioli,E.,Romero,I.A.,Wylezinska-

Arridge,M.,Couraud,P.O.,Lopez-Tremoleda,J.,Christian,H.C.,Weksler,

B.B.,etal.(2013).Identificationofanessentialendogenousregulatorof

blood-brainbarrierintegrity,anditspathologicalandtherapeuticimplications.

Proc.Natl.Acad.Sci.USA110,832–841.

deSousaEMelo,F.,Colak,S.,Buikhuisen,J.,Koster,J.,Cameron,K.,de

Jong,J.H.,Tuynman,J.B.,Prasetyanti,P.R.,Fessler,E.,vandenBergh,

S.P.,etal.(2011).Methylationofcancer-stem-cell-associatedWnttarget

genespredictspoorprognosisincolorectalcancerpatients.CellStemCell

9,476–485.

Ehrlicher,A.J.,Nakamura,F.,Hartwig,J.H.,Weitz,D.A.,andStossel,T.P.

(2011).MechanicalstraininactinnetworksregulatesFilGAPandintegrinbind-

ingtofilaminA.Nature478,260–263.

Elsir,T.,Smits,A.,Lindstro¨m,M.S.,andNiste′r,M.(2012).Transcriptionfactor

PROX1:itsroleindevelopmentandcancer.CancerMetastasisRev.31,

793–805.

Elyada,E.,Pribluda,A.,Goldstein,R.E.,Morgenstern,Y.,Brachya,G.,Cojo-

caru,G.,Snir-Alkalay,I.,Burstain,I.,Haffner-Krausz,R.,Jung,S.,etal.

(2011).CKIaablationhighlightsacriticalroleforp53ininvasivenesscontrol.

Nature470,409–413.

Fearon,E.R.(2011).Moleculargeneticsofcolorectalcancer.Annu.Rev.

Pathol.6,479–507.

Fodde,R.,andSmits,R.(2001).Diseasemodel:familialadenomatous

polyposis.TrendsMol.Med.7,369–373.

Gregorieff,A.,andClevers,H.(2005).Wntsignalingintheintestinalepithelium:

fromendodermtocancer.GenesDev.19,877–890.

Harvey,N.L.,Srinivasan,R.S.,Dillard,M.E.,Johnson,N.C.,Witte,M.H.,Boyd,

K.,Sleeman,M.W.,andOliver,G.(2005).Lymphaticvasculardefectspro-

motedbyProx1haploinsufficiencycauseadult-onsetobesity.Nat.Genet.

37,1072–1081.

Hu,Y.L.,DeLay,M.,Jahangiri,A.,Molinaro,A.M.,Rose,S.D.,Carbonell,W.S.,

andAghi,M.K.(2012).Hypoxia-inducedautophagypromotestumorcellsur-

vivalandadaptationtoantiangiogenictreatmentinglioblastoma.Cancer

Res.72,1773–1783.

Kemper,K.,Prasetyanti,P.R.,DeLau,W.,Rodermond,H.,Clevers,H.,and

Medema,J.P.(2012).MonoclonalantibodiesagainstLgr5identifyhuman

colorectalcancerstemcells.StemCells30,2378–2386.

Kozar,S.,Morrissey,E.,Nicholson,A.M.,vanderHeijden,M.,Zecchini,H.I.,

Kemp,R.,Tavare′,S.,Vermeulen,L.,andWinton,D.J.(2013).Continuous

clonallabelingrevealssmallnumbersoffunctionalstemcellsinintestinal

cryptsandadenomas.CellStemCell13,626–633.

Maschler,S.,Gebeshuber,C.A.,Wiedemann,E.M.,Alacakaptan,M.,

Schreiber,M.,Custic,I.,andBeug,H.(2010).AnnexinA1attenuatesEMT

andmetastaticpotentialinbreastcancer.EMBOMol.Med.2,401–414.

Merlos-Sua′rez,A.,Barriga,F.M.,Jung,P.,Iglesias,M.,Ce′spedes,M.V.,Ros-

sell,D.,Sevillano,M.,Hernando-Momblona,X.,daSilva-Diz,V.,Mun?oz,P.,

etal.(2011).Theintestinalstemcellsignatureidentifiescolorectalcancer

stemcellsandpredictsdiseaserelapse.CellStemCell8,511–524.

Metcalfe,C.,Kljavin,N.M.,Ybarra,R.,anddeSauvage,F.J.(2014).Lgr5stem

cellsareindispensableforradiation-inducedintestinalregeneration.CellStem

Cell14,149–159.

Myant,K.B.,Cammareri,P.,McGhee,E.J.,Ridgway,R.A.,Huels,D.J.,Cor-

dero,J.B.,Schwitalla,S.,Kalna,G.,Ogg,E.L.,Athineos,D.,etal.(2013).

ROSproductionandNF-kBactivationtriggeredbyRAC1facilitateWNT-

drivenintestinalstemcellproliferationandcolorectalcancerinitiation.Cell

StemCell12,761–773.

Parente,L.,andSolito,E.(2004).Annexin1:morethanananti-phospholipase

protein.Inflamm.Res.53,125–132.

Petrova,T.V.,Nyka¨nen,A.,Norrme′n,C.,Ivanov,K.I.,Andersson,L.C.,Haglund,

C.,Puolakkainen,P.,Wempe,F.,vonMelchner,H.,Gradwohl,G.,etal.(2008).

TranscriptionfactorPROX1inducescoloncancerprogressionbypromotingthe

transitionfrombenigntohighlydysplasticphenotype.CancerCell13,407–419.

Ragusa,S.,Cheng,J.,Ivanov,K.I.,Zangger,N.,Ceteci,F.,Bernier-Latmani,

J.,Milatos,S.,Joseph,J.-M.,Tercier,S.,Bouzourene,H.,etal.(2014).

PROX1PromotesMetabolicAdaptationandFuelsOutgrowthofWnt

high

MetastaticColonCancerCells.CellRep.PublishedonlineSeptember18,

2014.http://dx.doi.org/10.1016/j.celrep.2014.08.041.

Sadanandam,A.,Lyssiotis,C.A.,Homicsko,K.,Collisson,E.A.,Gibb,W.J.,

Wullschleger,S.,Ostos,L.C.,Lannon,W.A.,Grotzinger,C.,DelRio,M.,

etal.(2013).Acolorectalcancerclassificationsystemthatassociatescellular

phenotypeandresponsestotherapy.Nat.Med.19,619–625.

Sampieri,K.,andFodde,R.(2012).Cancerstemcellsandmetastasis.Semin.

CancerBiol.22,187–193.

Sansom,O.J.,Reed,K.R.,vandeWetering,M.,Muncan,V.,Winton,D.J.,

Clevers,H.,andClarke,A.R.(2005).CyclinD1isnotanimmediatetargetof

beta-cateninfollowingApclossintheintestine.J.Biol.Chem.280,28463–

28467.

Sato,K.,Tsuchihara,K.,Fujii,S.,Sugiyama,M.,Goya,T.,Atomi,Y.,Ueno,T.,

Ochiai,A.,andEsumi,H.(2007).Autophagyisactivatedincolorectalcancer

cellsandcontributestothetolerancetonutrientdeprivation.CancerRes.

67,9677–9684.

Sato,T.,Vries,R.G.,Snippert,H.J.,vandeWetering,M.,Barker,N.,Stange,

D.E.,vanEs,J.H.,Abo,A.,Kujala,P.,Peters,P.J.,andClevers,H.(2009).

SingleLgr5stemcellsbuildcrypt-villusstructuresinvitrowithoutamesen-

chymalniche.Nature459,262–265.

Sato,T.,Stange,D.E.,Ferrante,M.,Vries,R.G.,VanEs,J.H.,VandenBrink,S.,

VanHoudt,W.J.,Pronk,A.,VanGorp,J.,Siersema,P.D.,andClevers,H.(2011).

Long-termexpansionofepithelialorganoidsfromhumancolon,adenoma,

adenocarcinoma,andBarrett’sepithelium.Gastroenterology141,1762–1772.

Schepers,A.G.,Snippert,H.J.,Stange,D.E.,vandenBorn,M.,vanEs,J.H.,

vandeWetering,M.,andClevers,H.(2012).LineagetracingrevealsLgr5+

stemcellactivityinmouseintestinaladenomas.Science337,730–735.

Schwitalla,S.,Fingerle,A.A.,Cammareri,P.,Nebelsiek,T.,Go¨ktuna,S.I.,

Ziegler,P.K.,Canli,O.,Heijmans,J.,Huels,D.J.,Moreaux,G.,etal.(2013).

Intestinaltumorigenesisinitiatedbydedifferentiationandacquisitionof

stem-cell-likeproperties.Cell152,25–38.

Srinivasan,R.S.,Dillard,M.E.,Lagutin,O.V.,Lin,F.J.,Tsai,S.,Tsai,M.J.,

Samokhvalov,I.M.,andOliver,G.(2007).Lineagetracingdemonstratesthe

venousoriginofthemammalianlymphaticvasculature.GenesDev.21,

2422–2432.

Surrel,F.,Jemel,I.,Boilard,E.,Bollinger,J.G.,Payre′,C.,Mounier,C.M.,Tal-

vinen,K.A.,Laine,V.J.,Nevalainen,T.J.,Gelb,M.H.,andLambeau,G.(2009).

GroupXphospholipaseA2stimulatestheproliferationofcoloncancercellsby

producingvariouslipidmediators.Mol.Pharmacol.76,778–790.

VanderFlier,L.G.,Sabates-Bellver,J.,Oving,I.,Haegebarth,A.,DePalo,M.,

Anti,M.,VanGijn,M.E.,Suijkerbuijk,S.,VandeWetering,M.,Marra,G.,and

Clevers,H.(2007).TheIntestinalWnt/TCFSignature.Gastroenterology132,

628–632.

Wiener,Z.,Band,A.M.,Kallio,P.,Ho¨gstro¨m,J.,Hyvo¨nen,V.,Kaijalainen,S.,

Ritvos,O.,Haglund,C.,Kruuna,O.,Robine,S.,etal.(2014).Oncogenicmuta-

tionsinintestinaladenomasregulateBim-mediatedapoptosisinducedby

TGF-b.Proc.Natl.Acad.Sci.USA111,E2229–E2236.

Yeung,T.M.,Gandhi,S.C.,Wilding,J.L.,Muschel,R.,andBodmer,W.F.

(2010).Cancerstemcellsfromcolorectalcancer-derivedcelllines.Proc.

Natl.Acad.Sci.USA107,3722–3727.

Zheng,X.,Zhou,A.X.,Rouhi,P.,Uramoto,H.,Bore′n,J.,Cao,Y.,Pereira,T.,

Akyu¨rek,L.M.,andPoellinger,L.(2014).Hypoxia-inducedandcalpain-depen-

dentcleavageoffilaminAregulatesthehypoxicresponse.Proc.Natl.Acad.

Sci.USA111,2560–2565.

Zhou,A.X.,Hartwig,J.H.,andAkyu¨rek,L.M.(2010).Filaminsincellsignaling,

transcriptionandorgandevelopment.TrendsCellBiol.20,113–123.

1956CellReports8,1943–1956,September25,2014a2014TheAuthors

献花(0)
+1
(本文系一世秋缘首藏)