• 검색 결과가 없습니다.

Role of RIP3 in DNA damaging agent-induced breast cancer cell death

N/A
N/A
Protected

Academic year: 2021

Share "Role of RIP3 in DNA damaging agent-induced breast cancer cell death"

Copied!
48
0
0

로드 중.... (전체 텍스트 보기)

전체 글

(1)

저작자표시-비영리-변경금지 2.0 대한민국 이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게 l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다. 다음과 같은 조건을 따라야 합니다: l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건 을 명확하게 나타내어야 합니다. l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다. 저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다. 이것은 이용허락규약(Legal Code)을 이해하기 쉽게 요약한 것입니다. Disclaimer 저작자표시. 귀하는 원저작자를 표시하여야 합니다. 비영리. 귀하는 이 저작물을 영리 목적으로 이용할 수 없습니다. 변경금지. 귀하는 이 저작물을 개작, 변형 또는 가공할 수 없습니다.

(2)

Rol

eofRI

P3i

n DNA damagi

ng agent

-i

nduced

br

eastcancercel

ldeat

h

by

WooJung Ki

m

Maj

ori

n Mol

ecul

arMedi

ci

ne

Depar

t

mentofBi

omedi

calSci

ences

TheGr

aduat

eSchool

,Aj

ou Uni

ver

si

t

y

(3)

Rol

eofRI

P3i

n DNA damagi

ng agent

-i

nduced

br

eastcancercel

ldeat

h

by

WooJung Ki

m

A Di

s

s

e

r

t

a

t

i

onSubmi

t

t

e

dt

oTheGr

a

duat

eSc

hoolof

Aj

ouUni

ve

r

s

i

t

yi

nPa

r

t

i

alFul

f

i

l

l

me

ntoft

heRe

qui

r

e

me

nt

sf

or

TheDe

gr

e

eofMas

t

e

ro

fBi

omedi

calSci

ences

Super

vi

sedby

You-Sun Ki

m,Ph.

D.

Maj

ori

n Mol

ecul

arMedi

ci

ne

Depar

t

mentofBi

omedi

calSci

ences

TheGr

aduat

eSchool

,Aj

ou Uni

ver

si

t

y

(4)

Thi

scer

t

i

f

i

est

hatt

hedi

sser

t

at

i

on

ofWooJung Ki

m i

sappr

oved.

SUPERVI

SORY COMMI

TTEE

You-Sun Ki

m

Gyesoon Yoon

Seon-Yong J

eong

TheGr

aduat

eSchool

,Aj

ou Uni

ver

si

t

y

J

une,19t

h,2015

(5)

-ABSTRACT-Rol

eofRI

P3i

n DNA damagi

ng agent

-i

nduced

br

eastcancercel

ldeat

h

Receptor-interacting protein kinase 3 (RIP3 or RIPK3)is a central playerin “programmed”or“regulated”necroticcelldeath pathway.Herewe show thatprogrammed necrosisisactivated in responseto chemotherapeutic agents and contributes to chemotherapy-induced breast cancer celldeath. However,wealsoshow thatRIP3expression issilenced in basallikebreast cancercelllinesduetoDNA methylationnearitstranscriptionstartsite.Due to this silencing mechanism,loss ofRIP3 expression in these cells leads to resistance to DNA damaging-agents.Hypomethylating agents restore RIP3 expression and promote sensitivity notonly to death receptor ligands,but also to a surprising diversity of standard chemotherapeutic agents in a RIP3-specific manner.Ourdata indicates thatRIP3-dependentbreastcancer celldeath is activated in responseto DNA damaging-agents.Thissuggests thatRIP3playsagreaterroleinresponsetoDNA damaging-agentsthanhas been previously appreciated, we propose that hypomethylating agents, in combination with standard chemotherapy,may be usefulin treating breast cancersthatlackRIP3expression.

Keywords: RIP3, MLKL, Programmed necrosis, Hypomethylating agents, Chemotherapy

(6)

TABLE OF CONTENTS

ABSTRACT...i

TABLE OF CONTENTS ...iii LIST OF FIGURES ...v

I.INTRODUCTION ...1

II.MATERIALS AND METHODS ...4

A.Experimentalreagents...4

B.CellCulture...

.

.

.

.

.

...4

C.LentiviralshRNA experiments...4

D.ReverseTranscription-PCR...5

E.Cellviabilityassay...5

F.Westernblotanalysis...5

G.Immunoprecipitation...6

H.PyrosequencingofBisulfateconvertedgenomicDNA...6

I.Colonyformationassay...6

J.Invasionassay...7

III.RESULTS...8

A.RIP3expressionisoftensilencedinbreastcancercellsduetogenomic methylationnearitstranscriptionalstartsite...8

B.ExpressionofRIP3isessentialforTNF-inducednecroticcelldeath incancercells...13

C.RIP3expressionsensitizestoDNA-damagingagentsinbreastcancer cells...18 D.DNA-damagingagentsactivateRIP3-dependentMLKL

(7)

phosphorylation...24

E.RIP3actsastumorsuppressor...27

IV.DISCUSSION...31

REFERENCES...33

(8)

LI

ST OF FI

GURES

Fig.1.RIP3issilencedinmanybreastcancercelllines...10 Fig.2.HypomethylatingagentsrestoresRIP3expressionincancercells...11 Fig.3.RIP3issilencedbymethylationincancercells...12 Fig.4.ExpressionofRIP3isessentialforTNF-inducednecroticcelldeath..14 Fig.5.HypomethylatingagentrestoresTNF-inducednecroticcelldeathin

RIP3-deficientcells...15 Fig6.HypomethylatingagentdosenotaffectRIP3expressioninHT-29

cells...17 Fig.7.RestorationofRIP3byhypomethylatingagentenhancessensitivity

tochemotherapeuticagents...20 Fig.8.Hypomethylatingagent-inducedsensitizationtochemotherapeutic

inducednecroticcelldeathisdependentontheinductionofRIP3 expression...21 Fig.9.ExpressionofRIP3contributestosensitivitytoDNA-damaging

agents...22 Fig.10.DNA-damagingagentinducesnecrosomecomplexformation...23 Fig.11.DNA-damagingagentinducesMLKL phosphorylationinRI

P3-expressingcells...25 Fig.12.MLKL phosphorylationinducedbyDNA-damaging

isdependentonRIP3...26 Fig.13.RIP3suppressesmigrationandinvasioninbreastcancercells...29

(9)

I

.I

NTRODUCTI

ON

Resistance to celldeath is one hallmark ofcancercells,and tumorcells evolveavariety ofstrategiesto limitoreludeapoptosis.(Hanahan D etal., 2011).Evasion ofapoptosis may contribute to treatmentresistance and also tumor progression, since most current anticancer therapies including chemotherapy and immunotherapy primarily act by activating cell death pathwaysincluding apoptosisincancercells.Hence,abetterunderstandingof themolecularmechanismsunderlying tumorresistancetoapoptoticcelldeath isexpected toprovidethebasisforarationalapproach todevelopmolecular targetedtherapies.However,inadditiontoapoptosis,otherformsofcelldeath existprogrammednecrosis.

Programmed necrosis is activated in response to death receptor ligands andothertypeofcellularstressors(Declercqetal.,2009;Vandenabeeleetal., 2010;Vanlangenakkeretal..,2012b).Programmed or“regulated”necroticcell death is alternate programmed celldeath pathway that is different from apoptosis in that caspases are not essentialfor celldeath to occur,but necroticeventsleadtomembraneruptureandcelllysis(Galluzzietal..,2012), thus spilling the intracellular contents and ensuing the inflammation and immune system (N Vanlangenakkeratal.2012b;MichaelJ.Morgan,atal 2013).

Programmed celldeath can beinduced by aseveralstimuli,including the proinflammatory cytokine TNF-a.Studies ofthis mode ofcelldeath often utilize the murine fibrosarcoma cell line L929, or the human colon

(10)

adenocarcinomacelllineHT-29asmodels.In thecaseofL929cells,TNF-a prominently causes necrosis instead ofapoptosis,while in the case ofthe HT-29 cellline,the addition ofeitherSMAC mimetic orcycloheximide,in combinationwiththepancaspaseinhibitorzVAD,arenecessary toinducethis typeofdeath.

The kinase activity of Receptor-interacting protein kinase-3 (RIP3,or RIPK3) is an essentialfor the programmed necrotic celldeath execution (Duan-Wu Zhang et al.2009;Sudan He et al.2009;YoungSik Cho etal. 2009).Activation ofthecanonicalprogrammednecrosisincludestheformation ofa complex containing RIP3 and RIP1 (RIPK1)and recruitmentofmixed lineagekinasedomain-likeprotein (MLKL)(Zhao,Jetal2012;Sun,L etal 2012),whichisessentialfordeathtoproceed(Wu,Jetal2013;Murphy,Jet al2013).

RIP3-dependent phosphorylation and plasma membrane localization of MLKL isnecessary forprogrammednecroticcelldeathtooccur(Wang,H et al2014;Zhenyu Caietal2014;Chen,X etal2014).The translocation of MLKL to the membrane has been alternately reported to leads to plasma membranedisruption by through itsactivation ofion channelsorby directly permeabilizing theplasmamembrane(Wang,H etal2014;Dondelinger,Y et al2014).

In addition,programmed necrosisisnow acknowledged to play rolesin pathologicalprocesses,including a facilitativerolein tissuedamage,such as ischemia-reperfusion injury,andin hostdefenseofviralinfections.According to recentreportsthatprogrammed necrosiscould contributeto inflammatory skin diseases,chronicinflammation and high expression ofRIP3functionsin

(11)

inflammatorydiseasepathogenesis.

Here we show that effective cytotoxic chemotherapy is facilitated by RIP3-dependentprogrammed necroticcelldeath;however,RIP3expression is often silenced in cancer due to genomic methylation near the RIPK3 transcription startsite.We show a majority ofbreastcancercelllines and specially, basal like subtype lack RIP3 expression due to this silencing mechanism.Basallikesubtypeisonly foundinabout15~20percentofbreast cancer diagnoses.It has been shown to be aggressive,unresponsive to treatmentand,ultimately,indicativeofapoorprognosis.

LossofRIP3expression in basallikebreastcancercellsleadsto greater resistance not only to death receptor ligands, but also to a surprising diversityofchemotherapeuticagents.Treatmentofcellswithhypomethylating agents restore RIP3 expression and thereby promotes sensitivity to chemotherapeutics in a RIP3-specific manner,suggesting thatdeficiency of RIP3in tumorcellsisselected forduring tumordevelopmentand/orgrowth. Since hypomethylating agents are considerably well-tolerated in patients,an implication ofourstudy isthatRIP3-deficientbreastcancersubtypepatients maybenefitfrom receiving hypomethylating agentstoinduceRIP3expression priortotreatmentwithconventionalchemotherapeuticagents.

(12)

I

I

.MATERI

ALS AND METHODS

A.Experimentalreagents

TNF-α and zVAD were from R&D Systems (Minneapolis, MN). Anti-phopho-JNK(44682G), Anti-RIP3(72106) and phospho-MLKL(187091) antibodies were from Abcam (Cambridge, MA). Doxorubicin, etoposide, 5-fluorouracil, cisplatin, 5-AD and necrostatin-1 were purchased from Sigma-Aldrich (St. Louis, MO). cycloheximide were from Calbiochem (Danvers,MA).Dabrafenib was from Selleckchem (Houston,TX).SMAC mimetic(LCL-161)wasfrom AdooqBioscience(Irvine,CA).

B.CellCulture

Cancercelllines were cultured in recommended media from ATCC.All cellswereculturedat37℃ in5% CO2.HT-29andHeLacellswerecultured in DMEM supplemented with 10% fetalbovine serum,2mM glutamine,100 U/mlpenicillin,and 100 μg/mlstreptomycin.MDAMB231,HCC1937,SKBR3 and T47D cells werecultured in RPMIsupplemented with 10% fetalbovine serum,2mM glutamine,100U/mlpenicillin,and100μg/mlstreptomycin.

C.LentiviralshRNA experiments

Short-hairpin RNA (shRNA) plasmids targeting hRIP3 mRNA (NM_006871), and non-targeting control (NM_027088) were from Sigma-Aldrich.Lentiviralplasmids were transfected into 293T cells (System Biosciences, LV900A-1) using Lipofectamine 2000(Invitrogen). Pseudoviral

(13)

particles were collected 2 days afterthe lentiviralplasmid transfection and infected into cellswith polybrene(10 μg/mL).Infected cells werepuromycin selected2daysafterinfection.

D.ReverseTranscription-PCR

Reverse Transcription-PCR RNA was extracted using RNeasy (Qiagen). 200 ng oftotalRNA from each samplewasused forcDNA synthesiswith reversetranscriptase(Invitrogen).EqualamountsofcDNA productwereused forPCR with Taq DNA polymerase (Takara).Amplification was performed using thefollowing primers:RIP3sense(5′-CAAGGAGGGACAGAAATGGA -3′),RIP3antisense(5′-GCCTTCTTGCGAACCTACTG-3′),actinsense

(5′-CAGGTCATCACCATTGGCAATGAGC-3′),actinantisense(5′-GATG

TCCACGTCACACTTCATGA-3′)FinalPCR products wereresolved in 1%

agarosegelandstainedwithethidium bromide.

E.Cellviability assay

Representative images were taken by a phase-contrastmicroscope.Cell viability was determined using tetrazolium dye colorimetric tests (the MTT assay)readat570nm.Lactatedehydrogenaseleakagewasquantifiedusinga cytotoxicitydetectionkit(Promega,Madison,WI).

F.Western blotanalysis

Upon treatment,CellswerelysedinM2buffer(20mM TrisatpH 7,0.5%

NP-40,250mM NaCl,3mM EDTA,3mM EGTA,2mM DTT,0.5mM PMSF,

(14)

Equal cell extracts were resolved by 10% SDS-PAGE and analyzed by western blot using enhanced chemiluminescence (ECL, Amersham, Piscataway,NJ).

G.Immunoprecipitation

Cellslysiswasin M2buffer.Forimmunoprecipitation,lysatesweremixed andprecipitated with antibody andprotein G-agarosebeadsovernightat4℃. Boundproteinswereremovedbyboiling inSDS andresolvedbySDS-PAGE forimmunoblotting.

H.Pyrosequencing ofBisulfateconvertedgenomicDNA.

Pyrosequencing of RIPK3 Genomic DNA from cells was performed by Genomictree,Inc.Daejeon,South Korea using standard protocols.Briefly, Bisulfite-treated DNA samples were prepared using EZDNA Methylation-Lightning kit(Zymo Research)and then amplified by PCR.The reaction conditionswereasfollows:95℃ for10min → [95℃ for30sec→ 5 6℃ for 30sec → 72℃ for 30sec] 45cycles → 72℃ for 5min.From this reaction,2 μL of the PCR product was visualized by ethidium bromide staining on a 2% agarose gel.The remainder of the product underwent pyrosequencing performed on a PyroMark ID system (Qiagen) using a PyroGoldreagentkitforquantificationofthemethylationlevel.

I.Colony formation assay

T47D, SKBR3 cells were seeded in 6-well plates with a density of 100-500cellsperwellfor2–3weeks.Themedium wasdiscarded and each

(15)

wellwas washed twice with PBS carefully.The colonies were fixed in methanol for 10 min and then stained with Crystal Violet solution. Experimentswererepeatedthreetimes.

J.Invasion assay

Matrigel transwell invasion assay(BD Falcon #0006). Transparent PET membrane were coated with matrigel,and MDAMB231 cells were plated (2.5x105 cell/well) onto upper chamber in serum-free media and than put serum-add mediainto low chamber.Filterswereexcised,and migrated cells werestainedwithcrystalviolet.

(16)

I

I

I

.RESULTS

A.RI

P3expr

essi

on i

sof

t

en si

l

enced i

n br

eastcancercel

l

sduet

o

genomi

cmet

hyl

at

i

on neari

t

st

r

anscr

i

pt

i

onalst

ar

tsi

t

e

RIP3 is essentialforprogrammed necrosis (Duan-Wu Zhang etal.2009; Sudan He etal.2009;YoungSik Cho etal.2009).In the presentstudy,we examined RIP3 expression in subtypes ofbreastcancer by immunoblotting and RIP3 protein is clearly absent in basallike breast cancer,but other subtype or normal cells express RIP3 (Figure 1).To further test this hypothesis,RIP3expression isadjusted in breastcancercellsduetogenomic methylation so,we examined whetherDNA methylation was responsible for RIP3 deficiency. Treatment with the hypomethylating agent (5-aza-2'-deoxycytidine, abbreviated: 5-AD) restored RIP3 expression. In severalbreast cancer cells,5-AD induced RIP3 expression in a time or dose-dependentmanner(Figure2A-B).

Wefurtherexamined whetherDNA methylation wasresponsibleforRIP3 deficiency.First,we checked RIP3 expression cancercells or loss ofRIP3 cancercells by western blot(Figure 3A).To verify thatthe expression of RIP3wasassociatedwithmethylationofgenomicDNA nearthetranscription start site of RIP3 we conducted pyrosequencing of individual CpGs in bisulfate converted DNA within this region in RIP3 expression cells or unexpression cells.HT-29 and T47D cells,which express RIP3 highly,had very low methylation oftheentireregion.On theotherhand,had very low

(17)

methylation ofthe entire region (i.e.HCC1937,MDAMB231,HeLa)(Figure 3B).

ThesedatacollectivelydemonstratethattheRIP3transcriptionstartsiteis methylated in breastcancercelllinesand isnegatively associated with RIP3 expression.

(18)

Fig.1.RIP3issilencedin many breastcancercelllines.

Western blotting analysis oflysates from various breast cancer celllines showingRIP3expression.

(19)

A

B

Fig.2.Hypomethylating agentrestoresRIP3expression in cancercells. (A)Western blotting analysis ofRIP3 expression in HCC1937,MDAMB231, andHeLacellstreatedwith2μM 5-AD forindicatedtimepoints.

(B)Western blotting analysis ofRIP3 expression in HCC1937,MDAMB231, and HeLa celltreated with indicated concentrations (MDAMB231 and HeLa cellsfor4days,HCC1937cellfor8days.)

(20)

A

B

Fig.3.RIP3issilencedby methylation in cancercelllines.

(A)Western blotting analysisofRIP3expression in HT-29,T47D,HCC1937, MDAMB231 and HeLa cells.(B) Summary of pyrosequencing analysis of bisulfite-converted genomicDNA from T47D,HT-29,HCC1937,MDAMB468, andHeLacells.

(21)

B.

Expr

essi

on ofRI

P3 i

sessent

i

alf

orTNF-i

nduced necr

ot

i

ccel

l

deat

h i

n cancercel

l

s.

RIP3 isessentialmolecularforprogrammed necrosis (Duan-Wu Zhang et al. 2009; Sudan He et al. 2009; YoungSik Cho etal. 2009). First, we experimented with HT-29 celland HeLa cellwhich were researched about necroptosis.Consistentwith the many paper,cells deficientRIP3 expression are completely resistantto prototypicalprogrammed necroticstimuli(TNF-α+ zVAD + eitherSMAC mimetic orcycloheximide;also referred to herein as TSZ or TCZ), so become sensitive when RIP3 is ectopically expressed (Figure 4A) and then cell viability was analyzed by phase-contrast microscopy(Figure4B).

We nextsoughtto testwhetherhypomethylating agents would sensitize RIP3-deficientcellstonecropticcondition (TSZ,TCZ)viarestoration ofRIP3 expression.Treatmentwith thehypomethylating agent(5-AD)restored RIP3 expression in HeLa cells (Figure 5A). TNF-α induced necrosis become sensitive after 5-AD treatment (Figure 5B).RIPK1 inhibitor necrostatin-1 blocks cell death upon 5-AD treatment followed by necrotic stimulus, indicating thecanonicalregulated necroticdeath pathway isactivated(Figure 5C).Importantly,5-AD does notaffectnecrotic celldeath in HT-29 cells (Figure 6),suggesting the sensitizing effect of 5-AD is mediated though inductionofRIP3expression.

(22)

A B

Fig.4.Expression ofRIP3isessentialforTNF-inducednecroticcell death.

(A)Viability (MTT)ofHeLacellectopically expressing RIP3treated 36hrs with TCZ [TNF (30ng/mL),cycloheximide(CHX,2μg/mL),and zVAD (20 μM)]orTSZ [TNF,SMAC mimetic(100nM),and zVAD].(B)Cellviability wasanalyzedbyphase-contrastmicroscopy.

(23)

A

(24)

Fig.5.Hypomethylating agentrestoresTNF-inducednecroticcelldeath in RIP3-deficientcells.

(A)HeLacellsweretreatedwith 5-AD (2μM,4day)andcelllysateswere analyzed by immunoblotting and reverse transcription-PCR.(B) HeLa cells werepretreatedwith5-AD (2μM,4day)andthenfurthertreatedwithTCZ [TNF (30ng/mL),cycloheximide(CHX,0.5,1,2μg/mL),andzVAD (20μM)] for48hrs.(C)TNF-induced necroticcelldeath in RIP3-restored HeLacells is inhibited by necrostatin-1.Cells were pretreated with necrostatin-1 (40 μ M) for 1 h before TCZ or TSZ treatment for 48 hrs and cellviability analyzedbyMTT assay.

(25)

Fig. 6. Hypomethylating agent dose not affect RIP3 expression in HT-29cells.

HT-29 cells were pretreated with 5-AD (2 μM,4 day) and then further treatedwithTCZ[TNF (30ng/mL),cycloheximide(CHX,1,2,5μg/mL),and zVAD (20μM)]for48hrsandcellviabilityanalyzedbyMTT assay.

(26)

C.RI

P3 expr

essi

on sensi

t

i

zes t

o DNA-damagi

ng agent

si

n br

east

cancercel

l

s

Wethereforesoughttodeterminewhetherhypomethylating agentspromote chemotherapeutic cell death by via enhancing RIP3 expression. First, hypomethylating agent would sensitize RIP3-deficient cells to necroptic condition (TSZ,TCZ)viarestoration ofRIP3expression.Treatmentwith the hypomethylating agent(5-AD)restored RIP3expression in MDAMB231cells (Figure7A-B)

Indeed, 5-AD treatment sensitizes MDAMB231 cells to doxorubicin (Doxo),etoposide (Etopo),5-Fluorouracil(5-FU),Camptothecin (CPT),and Cisplatin (Figure 7C),suggesting induction ofRIP3 may contribute to cell death from a wide range of chemotherapeutic drugs with different mechanisms ofaction.In the absence ofRIP3,hypomethylating agents lose their ability to sensitize cells to chemotherapeutic agents (Figure 8), confirming thattheirsensitization isdependenton increased RIP3expression. While the above data indicate that programmed necrosis contributes to chemotherapeuticcytotoxicity.

We examined whether RIP3 contributed to death from chemotherapeutic agents,a role forRIP3 in celldeath induced by standard chemotherapeutic cytotoxic agents has neverbeen reported.In MDAMB231 cells (which lack endogenous RIP3 expression),MDAMB231 cellectopically expressing RIP3 treated necroptic condition (TSZ,TCZ)and cellviability was analyzed by immunoblotting (Frigure 9A-B).The ectopic expression of RIP3 bestowed additionalsensitivitybothtoetoposide,doxorubicinandCPT asmeasuredby

(27)

MTT assays (Figure 9C). suggesting that RIP3 can contribute to the cytotoxicityofmultipledrugswithdiversemechanismsofaction.

Wesoughttoinvestigatethemechanism bywhichRIP3wassensitizing to chemotherapeutics.Weexamined whetherRIP3wasin thesamecomplex as caspase-8 upon treatment of cells with doxorubicin. Doxorubicin led to interaction ofcaspase-8 with RIP1 and RIP3,along with FADD,though no interaction was detected in RIP3 knockdown HT-29 cells (Figure 10).this data suggested to us that programmed necrosis might be important for RIP3-dependentchemocytotoxicity.

(28)

A

B C

Fig. 7. Restoration of RIP3 by hypomethylating agent enhances sensitivity tochemotherapeuticagents.

(A)MDAMB231cellsweretreatedwith thehypomethylating agent,5-AD (2 μM,4day)andthen analyzedRIP3expression by western blotting. (B)Cells from (A)weretreatedwithTCZ [TNF (30ng/mL),cycloheximide(CHX,2μ g/mL),and zVAD (20 μM)]for48 hrs and cellviability analyzed by MTT assay.(C)MDAMB231cellsweretreatedwith5-AD (2μM,4day)andthen furthertreated with camptothecin (CPT μg/mL),cisplatin (μM),doxorubicin (DoxoμM),etoposide(EtopoμM),or5-fluorouracil(5-FU,μM)for36hours. CellviabilitywasanalyzedbyMTT assay.

(29)

Fig.8.Hypomethylating agent-induced sensitization to chemotherapeutic induced necrotic cell death is dependent on the induction of RIP3 expression.

MDAMB231 cells were infected with lentivirus encoding RIP3 shRNA ora non-silencing control.After selection with puromycin,cells were pretreated with 5-AD (2μM)for4 day and then analyzed forknockdown by western blotting or treated with doxorubicin (μM),etoposide (μM),camptothecin (μ g/mL)for48hrsandcellviabilitythenanalyzedbyMTT assay.

(30)

A

B C

Fig.9.Expression ofRIP3 contributesto sensitivity to DNA-damaging agents.

(A)MDAMD231cellswereinfected with pLX303-hRIP3lentiviralplasmid to establish stable RIP3-expressing cells.RIP3 expression was confirmed by western blot. (B-C) MDAMB231 cells ectopically expressing RIP3 were treated with TCZ [TNF (30 ng/mL),cycloheximide (CHX,2 μg/mL),and zVAD (20 μM)]for48 hrs and cellviability was analyzed by MTT assay. (C)Cells were treated with indicated concentration ofdoxorubicin,etoposide orCPT for2dayandcellviabilitywasanalyzedbyMTT assay.

(31)

Fig.10.Doxorubicin inducesnecrosomecomplex formation.

Western blotting of Caspase-8 immunoprecipitates from HT-29 and RIP3 knockdown HT-29 cells pretreated with zVAD (20 μM)for1 hr,and then treatedwithdoxorubicin(5μM)for18hrs.

(32)

D. DNA-damagi

ng agent

s act

i

vat

e RI

P3-dependent MLKL

phosphor

yl

at

i

on

RIP3-dependent phosphorylation and subsequent plasma membrane localization ofMLKL isnecessary forprogrammednecroticcelldeath (Caiet al.,2014;Chen etal.,2014).WeexaminedwhetherRIP3dependentofMLKL activationby doxorubicin.DoxorubicininducedtimeordosedependentMLKL phosphorylationinendogenousRIP3expressionT47D cells(Fig11A-B).

We examined whether RIP3 contributed to death from chemotherapeutic agents in ectopically expressing RIP3 MDAMB231 and HCC1937 cells. Phosphorylated MLKL was detected in MDAMB231 and HCC1937 cells treated with doxorubicin (Figure 12A-B),suggesting thatRIP3 potentiates these death processes through activation of MLKL-dependent programmed necrosis.indicating thatMLKL activation contributedtodoxorubicin-mediated celldeath.

(33)

A B

Fig. 11. DNA-damaging agent induced MLKL phosphorylation in

RIP3-expressing cells.

(A) T47D cells were treated with doxorubicin (μM) for 24 hrs and cell lysateswereanalyzedby western blotting.(B)Western blotsoflysatesfrom T47D cellstreatedwithDoxorubicin(2μM)atindicatedtimepoints.

(34)

A B

Fig.12.MLKL phosphorylation induced by DNA-damaging agent is

dependenton RIP3.

(A) Western blots of lysates from Mock vector and RIP3 overexpressing MDAMB231cellstreatedwithdoxorubicin(2μM)atindicatedtimepoints.(B) RIP3 expressing cells were treated with different concentrations of doxorubicinfor24hrsandcelllysateswereanalyzedbywesternblotting.

(35)

E.RI

P3act

sasat

umorsuppr

essor

WhilelossofgenesinvolvedinDNA repaircanrendercancercellsmore susceptible to DNA-damaging chemotherapies, many are also tumor suppressors because theirloss results in an increase in genomic instability and oncogenic growth.Therefore,we tested the hypothesis that RIP3 is acting asatumorsuppressor,anditslosswillpromotemalignantphenotypes, including increased growth rates, invasive potential. We performed these experimentsinMDAMB231,T47D andSKBR3cells.Thesedifferentcelllines were chosen because they representa sampling ofdifferentbreastcancer types.MDAMB231 cells are triple negative (ER-,PR-,HER2) and have completed epithelialto mesenchymaltransition (EMT)(Dawn A etal,1999, HonorHugoetal,2007),T47D and SKBR-3cellsoverexpressHER2and do notexpressER orPR.

To test whether expression of RIP3 could increase breast cancer cell migration,we performed scratch-wound migration assays.MDAMB231 cell wereinfected with RIP3orcontrollentivirusand assayed fortheirability to filla scratch wound.As in the wound healing assays,the MDAMB231 cell moreslowlyfilledthewoundwhenincreasedforRIP3-overexpressing(Figure 13A).However,theresults from thescratch-wound assay could also reflect increased cell proliferation, so we additionally measured the effect of RIP3-overexpressing on cellinvasion.In contrastto the results observed above, RIP3-overexpressing significantly decreased the invasion of MDAMB231cell(Figure13B).

(36)

formation assay.T47D andSKBR3celllineswereinfectedwith RIP3KD or control lentivirus. RIP3 knockdown cells more quickly filled the colony formation (Figure 13C),and in anothercelllines also showed same results (Figure13D).ThesedatasuggestthatRIP3haveatumorsuppressorfunction andwillplayakeyroleintumorigenesis.

(37)

A B

(38)

Fig.13.RIP3suppressesmigration andinvasion in breastcancercells. (A) Scratch wound healing assay in mock vector MDAMB231 and RIP3 overexpressing MDAMB231 cells. Wound healing was analyzed by phase-contrastmicroscopy. (B)Matrigeltranswellinvasion assay.Transwell filters werecoated with matrigeland MDAMB231 cells wereplated (2.5x105 cell/well)ontoupperchamberinserum-freemediaandthanputserum-added media into low chamber.Filters were stained with crystalviolet.(C)T47D and SKBR3 cells were infected with a lentivirus encoding RIP3 shRNAs or non-silencing control vector. After selection with puromycin, cells were analyzed forknockdown by western blotting.Cellgrowth was analyzed by colony formation assay after15days.(D)RIP3knock-downedHT-29cellor RIP3 overexpressing HeLa cellgrowth was analyzed by colony formation assayafter15days.

(39)

I

V.DI

SCUSSI

ON

Programmed necrosis has been identified as an important mechanism underlying multiple physiologicaland pathologicalprocesses (Vanlangenakker et al.,2012a;Kaczmarek et al.,2013).Here we have discussed data that indicatesthattheexpression ofRIP3islostin breastcancercells,and that the mechanism forthis silencing is largely due to DNA methylation ofthe RIPK3genomicsequence.RIP3expression may berestored in a majority of cells with RIP3 deficiency by treating them with hypomethylating agents, thusrestoringsensitivityofthesecellstoprogrammednecrosis.

Our data indicate thatRIP3 expression is likely selected againstduring cancergrowth orprogression,with methylation ofthe genomic region near theRIPK3transcription startsiteleading toRIP3silencing.SinceRIP3plays a greater role in response to chemotherapeutics than has been previously appreciated.The response ofcells to 5-AD suggeststhe exciting possibility that RIP3 expression may be manipulated in cancer cells to make them sensitive to regulated necrosis,we propose thathypomethylation agents,in combination with standard chemotherapy,may be usefulin treating cancers that lack RIP3 expression. RIP3 deficiency is likely to have important biological and therapeutic consequences,including possible effects on the responses oftumorcells to chemotherapy.How RIP3 is activated by from drugs with such mechanistic differences is an importantmatter for future investigation and wewillinvestigatehow thetumorsuppressorrolein RIP3

haveanymechanism.

(40)

treatment of cancer by potentially restoring RIP3 expression in human patients.

(41)

REFERENCES

1.Andrew Oberst, Christopher P. Dillon, Ricardo Weinlich, Laura L. McCormick,PatrickFitzgerald,CristinaPop,RazqHakem,Guy S.Salvesen & DouglasR.Green.“Catalyticactivity ofthecaspase-8-FLIP(L)complex inhibitsRIPK3-dependentnecrosis.”Nature 471(7338):363-367,2011

2.Bonnet,M.C.Preukschat,D,Welz,P.S,van Loo,G,Ermolaeva,M.A, Bloch,W & Pasparakis,M.“TheadaptorproteinFADD protectsepidermal keratinocytes from necroptosis in vivo and prevents skin inflammation.”

Immunity,35(4),572-582.2011

3.Chen,X.,Li,W.,Ren,J.,Huang,D.,He,W.T.,Song,Y.,...& Han,J. “Translocation of mixed lineage kinase domain-like protein to plasma membraneleadstonecroticcelldeath.”Cellresearch,24(1),105-121,2014 4.Dondelinger,Y.,Declercq,W.,Montessuit,S.,Roelandt,R.,Goncalves,A.,

Bruggeman,I.& Vandenabeele,P.“MLKL compromisesplasmamembrane integrity by binding tophosphatidylinositolphosphates.”Cellreports,7(4), 971-981,2014

5.Dawn A.Kirschmann,Elisabeth A.Seftor,DanielR.C.Nieva,Elpidio A. Mariano,MaryJ.C.Hendrix.“Differentiallyexpressedgenesassociatedwith the metastatic phenotype in breast cancer.Breast cancer research and treatment.”55:127-136,1999

6.Duan-Wu Zhang,Jing Shao,Juan Lin,NaZhang,Bao-Ju Lu, Sheng-Cai

Lin,Meng-Qiu Dong,JiahuaiHan.“RIP3,an energy metabolism regulator thatswitches TNFinduced celldeath from apoptosis to necrosis.”Science

(42)

7.French,L.E.“TNF-α and IFN-γ are potentialinducers ofFas-mediated keratinocyteapoptosisthrough activation ofinduciblenitricoxidesynthase in toxic epidermal necrolysis.” Journal of Investigative Dermatology,

133(2),489-498,2013

8.Galluzzi,L.,Vitale,I.,Abrams,J.M.,Alnemri,E.S.,Baehrecke,E.H., Blagosklonny,M.V.& Kroemer,G.“Molecular definitions of celldeath subroutines:recommendationsoftheNomenclatureCommitteeonCellDeat 2012.”CellDeath& Differentiation,19(1),107-120,2012

9.Gunther, C, Martini, E, Wittkopf, N, et al. “Caspase-8 regulates TNF-alpha-induced epithelialnecroptosis and terminalileitis.”Nature 477

(7364):335-U108,2011

10.Hildebrand,J.M.,Tanzer,M.C.,Lucet,I.S.,Young,S.N.,Spall,S.K., Sharma,P.& Silke,J.“Activation ofthe pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptoticcelldeath.”ProceedingsoftheNationalAcademyofSciences, 111(42),15072-15077,2014

11.HonorHugo,M.Leigh Ackland,Tony Blick,MitchellG.Lawrence,Judith A.Clements,Elizabeth D.Williams,and Erik W.Thompson.“Epithelial --mesenchymal and mesenchymal—epithelial transitions in carcinoma progression.”Journalofcellularphysiology.213:374-383,2007

12.Hanahan,D,Weinberg,RA.“HallmarksofCancer:TheNextGeneration.”

Cell144(5):646-674,2011

13.Kaiser,W.J.,Upton,J.W.,& Mocarski,E.S.“Viralmodulation of programmednecrosis.”Currentopinioninvirology,3(3),296-306,2013

(43)

14.Linkermann,A,Bräsen,JH,Darding,M,Jin,M.K,Sanz,A.B,Heller,J. O & Krautwald,S.“Two independent pathways of regulated necrosis mediate ischemia–reperfusion injury.” Proceedings of the National AcademyofSciences,110(29),12024-12029,2013

15.Lu,J.V.,Weist,B.M.,van Raam,B.J.,Marro,B.S.,Nguyen,L.V., Srinivas,P.& Walsh,C.M.“Complementary roles of Fas-associated death domain (FADD)and receptorinteracting protein kinase-3 (RIPK3) in T-cell homeostasis and antiviral immunity.” Proceedings of the NationalAcademyofSciences,108(37),15312-15317.2011

16.Moriwaki,K,Chan,FKM.“RIP3:a molecular switch for necrosis and inflammation.”Genes& Development27(15):1640-1649,2013

17.Murphy,J.M.,Czabotar,P.E.,Hildebrand,J.M.,Lucet,I.S.,Zhang,J. G., Alvarez-Diaz, S. & Alexander, W. S. “The pseudokinase MLKL mediatesnecroptosisviaamolecularswitch mechanism.”Immunity,39(3), 443-453.2013

18.MichaelJ.Morgan, Zheng-gang Liu. “Programmed celldeath with a necrotic-likephenotype.”BioMolecularConcepts4(3):259-275,2013

19.N Vanlangenakker,T Vanden BergheandP Vandenabeele.“Many stimuli pullthenecrotictrigger,anoverview.”CellDeathDiffer19:75.86,2012b

20.Peter Vandenabeele, Lorenzo Galluzzi, Tom Vanden Berghe & Guido Kroemer. “Molecular mechanisms of necroptosis: an ordered cellular explosion.”NatRevMolCellBiol11:700–14,2010

21.Sudan He,LaiWang,Lin Miao,Tao Wang,Fenghe Du,Liping Zhao,

(44)

necroticresponsetoTNF-alpha.”Cell137(6):1100-1111,2009

22.Sun,L.,Wang,H.,Wang,Z.,He,S.,Chen,S.,Liao,D.& Wang,X.

“Mixed lineage kinase domain-like protein mediates necrosis signaling downstream ofRIP3kinase.”Cell,148(1),213-227,2012

23.Viard-Leveugle, I., Gaide, O., Jankovic, D., Feldmeyer, L., Kerl, K., Pickard,C.& Galluzzi,L.,Vitale,I.,Abrams,J.M.,Alnemri,E.S., Baehrecke, E. H., Blagosklonny, M. V.& Kroemer, G. “Molecular definitionsofcelldeathsubroutines:recommendationsoftheNomenclature Committee on CellDeath.”CellDeath & Differentiation,19(1),107-120, 2012

24.Wim Declercq,Tom Vanden Berghe,PeterVandenabeele.“RIP Kinasesat theCrossroadsofCellDeathandSurvival.”Cell138:229.32,2009

25.William J. Kaiser, Jason W. Upton, Alyssa B. Long, Devon Livingston-Rosanoff,LisaP.Daley-Bauer,Razqallah Hakem, Tamara Caspary& EdwardS.Mocarski.“RIP3mediatestheembryoniclethalityof caspase-8-deficientmice.”Nature471(7338):368-372.2011

26.Welz,P.S,Wullaert,A,Vlantis,K.,Kondylis,V,Fernández-Majada,V, Ermolaeva,M,& Pasparakis,M.“FADD preventsRIP3-mediatedepithelial cell necrosis and chronic intestinal inflammation.” Nature, 477(7364), 330-334,(2011).

27.Wang,H.,Sun,L.,Su,L.,Rizo,J.,Liu,L.,Wang,L.F.& Wang,X.

“Mixed lineage kinase domain-like protein MLKL causes necrotic membranedisruptionuponphosphorylationbyRIP3.”Molecularcell,54(1), 133-146,2014

(45)

28.Wu,J.,Huang,Z.,Ren,J.,Zhang,Z.,He,P.,Li,Y & Han,J. “Mlkl knockoutmicedemonstratetheindispensableroleofMlklin necroptosis.”

Cellresearch,23(8),994-1006,2013

29.Xin Chen1, Wenjuan Li1, Junming Ren1, Deli Huang, Wan-ting He, Yunlong Song,Chao Yang,Wanyun Li,Xinru Zheng1,Pengda Chen and JiahuaiHan. “Translocation ofmixed lineagekinase domain like protein to plasma membrane leads to necrotic celldeath.” CellRes.24:105–21, 2014

30.YoungSik Cho,Sreerupa Challa,David Moquin,Ryan Genga,Tathagat

Dutta Ray, Melissa Guildford, Francis Ka-Ming Chan.

“Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation.” Cell 137 (6):1112-1123,2009

31.You-Sun Kim, Michael J. Morgan, Swati Choksi, Zheng-gang Liu. “TNF-inducedactivation oftheNox1NADPH oxidaseand itsrolein the inductionofnecroticcelldeath.”MolCell26:675–87,2007

32.Y-T Wu, H-L Tan, Q Huang, X-J Sun, X Zhu and H-M Shen.

“zVAD-induced necroptosisin L929cellsdependson autocrineproduction ofTNFalpha mediated by the PKCMAPKs- AP-1 pathway.”CellDeath Differ18:26–37,2011

33.Zhenyu Cai, Siriporn Jitkaew, Jie Zhao, Hsueh-Cheng Chiang, Swati Choksi,JieLiu,YvonaWard,Ling-gangWu& Zheng-Gang Liu.“Plasma membrane translocation of trimerized MLKL protein is required for TNF-inducednecroptosis.”NatCellBiol.16:55–65,2014

(46)

34.Zhang,H.,Zhou,X.,McQuade,T.,Li,J.,Chan,F.K.M.,& Zhang,J. “Functionalcomplementation between FADD and RIP1 in embryos and lymphocytes”.Nature,471(7338),373-376,2011

35.Zhao,J.,Jitkaew,S.,Cai,Z.,Choksi,S.,Li,Q.,Luo,J.,& Liu,Z.G. “Mixed lineagekinasedomain-likeisakey receptorinteracting protein 3 downstream component of TNF-induced necrosis.” Proceedings ofthe NationalAcademyofSciences,201200012,2012

(47)

국문요약

-DNA damagi

ng agent

s에 의한 유방암 세포 사멸에서

RI

P3의 역할에 관한 연구

아주대학교 대학원 의생명과학과 분자의학전공 김 우 중 (지도교수 :김 유 선) 수용체- 상호 작용 단백질 키나제 3(Receptor-interacting protein kinase-3) 는 ‘프로그램’또는 ‘조절되는’괴사 세포사멸에서 중요한 역할을 하는 단백질이 다.본 연구에서,화학치료제가 프로그램된 괴사를 활성화시키고,화학치료에 의 한 유방암세포 사멸에 기여함을 보여주었다.하지만,RIP3발현이 전사 시작 지

점 근처의 DNA 메틸화에 의해 basallike유방암세포에서 억제되어 있음을 확인

하였다.이러한 억제 기전을 통한,basallike유방암 세포에서의 RIP3발현 손실

은 DNA 손상 약물에 대한 저항성을 야기한다.반면,탈메틸화제는 RIP3의 발현 을 회복시키고 사멸 수용체 리간드에 대한 감수성 뿐만 아니라 놀랍도록 다양한 표준 화학치료 약물에 대한 감수성 역시 RIP3 특이적으로 촉진하였다.본 결과 에서는 RIP3의존적 유방암 세포사멸이 DNA 손상 약물에 반응하여 활성화됨을 확인하였다.이러한 결과는 RIP3가 이전에 인식 된 것보다 DNA 손상 약물에 대 한 반응에서 중요한 역할을 함을 보여주었으며,표준 화학치료와 함께 탈메틸화 제를 병합 처리하는 것이 RIP3가 발현하지 않는 유방암 치료에 유용한 치료법이 될 것이라고 제안한다.

(48)

참조

관련 문서

Effect of LRP6 on survivin expression in hypoxic cardiomyocytes (A) Expression of survivin in Ad-LRP6 and silencing LRP6 infected cardiomyocytes as determined by western

These results suggest that the bilobalide inhibits the cell proliferation and induces the apoptotic cell death in FaDu human pharyngeal squamous cell carcinoma via both

whether NME1-depleted cells impairs the Rad51 foci formation in response

1 1, 3 3 Sc he i nman등 34 은 경동맥에 손상을 받은 쥐에 p53을 활성화시킴으로써 조직의 세포고사가 증가함을 보고하였다.즉 p53 의 단백 발현 정도를 통해 DNA 손상

마지막으로 남녀 중학생들의 무용 표현활동 에 대한 능력믿음은 가치지각 , ( ) 을 통해서 뿐만 아니라 직접적으로 수업참여 수준에 긍정적인 영향을 미치는 지를

DNA 프로브법은 기존의 세균배양법, 생화학검사법 및 DNA-DNA hybri di zati on법 등에 비해 재현성,신속성 및 경제적 측면에서 뛰어나다.그러나 중합

Activated H-Ras expression in human fibroblast cell lines increases the activity of Ku80 to bind injuried DNA, reduces γ-H2AX expression by UV irradiation,

Glutamate excitotoxicity induced by excessive activation of NMDA receptor causes various damage to cells, which leads to cell death.. In previous studies, increased ROS