• Keine Ergebnisse gefunden

The ubiquitin-like modifier FAT10 in cancer development

N/A
N/A
Protected

Academic year: 2022

Aktie "The ubiquitin-like modifier FAT10 in cancer development"

Copied!
11
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

The ubiquitin-like modifier FAT10 in cancer development

Annette Aichem

a

, Marcus Groettrup

a,b,∗

aBiotechnologyInstituteThurgauattheUniversityofKonstanz,CH-8280Kreuzlingen,Switzerland

bDivisionofImmunology,DepartmentofBiology,UniversityofKonstanz,D-78457Konstanz,Germany

a b s t r a c t

Duringthelastyearsithasemergedthattheubiquitin-likemodifierFAT10isdirectlyinvolvedincancer development.FAT10expressionishighlyup-regulatedbypro-inflammatorycytokinesIFN-␥andTNF-␣ inallcelltypesandtissuesanditwasalsofoundtobeup-regulatedinmanycancertypessuchasglioma, colorectal,liverorgastriccancer.Whilepro-inflammatorycytokineswithinthetumormicroenvironment probablycontributetoFAT10overexpression,anincreasingbodyofevidencearguesthatpro-malignant capacitiesofFAT10itselflargelyunderlieitsbroadandintenseoverexpressionintumortissues.FAT10 therebyregulatespathwaysinvolvedincancerdevelopmentsuchastheNF-␬B-orWnt-signaling.More- over,FAT10directlyinteractswithandinfluencesdownstreamtargetssuchasMAD2,p53or␤-catenin, leadingtoenhancedsurvival,proliferation,invasionandmetastasisformationofcancercellsbutalsoof non-malignantcells.InthisreviewwewillprovideanoverviewoftheregulationofFAT10expressionas wellasitsfunctionincarcinogenesis.

Contents

1. Introduction...452

2. Theubiquitin-likemodifierFAT10,aproteinoftheimmunesystem...452

2.1. ThestructureofFAT10andhowittargetsforproteasomaldegradation...452

2.2. FAT10inadaptiveandinnateimmunity...452

2.3. WhicharethetargetsofFAT10andhowisitconjugatedtoitssubstrates...453

3. RegulationofFAT10expressionunderinflammatoryconditionsandduringcancerdevelopment...454

3.1. FAT10expressionlevelsindifferentcancertypes...454

3.2. ThetransformingcapacityofFAT10...454

4. WhatarethemechanismsbehindFAT10overexpression?...455

5. FunctionalconsequencesofFAT10up-regulationininflammationandcancer ... 455

5.1. MAD2andchromosomalinstability...455

5.2. p53...456

5.3. ␤-catenin...458

5.4. Smad2...459

6. Summaryandfutureprospects...459

Conflictofinterest ... 459

Acknowledgements...459

References...459

Abbreviations:AIPL1,aryl-hydrocarbonreceptorlike-1;APC/C,anaphase-promotingcomplex/cyclosome;DUB,deubiquitylatingenzyme;EMT,epithelial-mesenchymal transition;FAT10,HLA-Fadjacenttranscript10;GSK,glycogensynthasekinase;HCC,hepatocellularcarcinoma;HLA,humanleukocyteantigen;HOX,homeobox;IFN, interferon;IL,interleukin;MAD2,mitoticarrestdeficient2;mTEC,medullarythymicepithelialcells;NF-␬B,nuclearfactorkappa-B;NUB1L,NEDD8-ultimatebuster1long;

STAT,signaltransducerandactivatoroftranscription;SUMO,small-ubiquitin-likemodifier;TGF,transforminggrowthfactor;TNF,tumornecrosisfactor;UBD,UbiquitinD;

ULM,ubiquitin-likemodifier;UPS,ubiquitinproteasomesystem;USE1,UBA6-specificE2enzyme1.

Correspondingauthorat:DivisionofImmunology,DepartmentofBiology,UniversityofKonstanz,Universitaetsstr.10,D-78457Konstanz,Germany.

E-mailaddress:marcus.groettrup@uni-konstanz.de(M.Groettrup).

Konstanzer Online-Publikations-System (KOPS) URL: http://nbn-resolving.de/urn:nbn:de:bsz:352-0-347923

Erschienen in: The International Journal of Biochemistry & Cell Biology ; 79 (2016). - S. 451-461 https://dx.doi.org/10.1016/j.biocel.2016.07.001

(2)

1. Introduction

Acausallinkbetweenchronicinflammationorinfectionand tumorigenesisisnowadaysagenerallyacceptedprincipleleading tothedevelopmentofvariouscancertypes.Theunderlyingmech- anismisinpartbasedontheupregulationandsecretionofseveral proinflammatorycytokinessuchastumornecrosisfactor(TNF)␣, interferon(IFN)␥orinterleukin-6(IL-6)bystromalcells aswell asbyinfiltratingimmunecells inthetumormicroenvironment, orby cytokines,secreted bytumor cells themselves(Zamarron andChen,2011).Suchinflammatoryconditionsaregeneratedto promote healing and regeneration of the affected tissue. How- ever,thepersistingexpressionofthesecytokinescanalsoprovide anicheforcancerdevelopmentbypromotingDNAdamageand abnormal tissue healing. This is based on the fact that pro- inflammatory cytokines cause the activation of transcription factorssuchasnuclearfactorkappa-B(NF-␬B)andsignaltrans- ducerandactivatoroftranscription(STAT)3,whichplayacentral roleasactivatorsofanti-apoptoticgeneexpressionandcellprolif- eration.Furthermore,thesecytokinespromoteoptimalconditions for tumorcell proliferation,invasion and metastasisformation, apoptosisresistanceaswellasangiogenesis,i.e.processeswhich areallprerequisitesforcancerdevelopment(Grivennikovetal., 2010;Karin,2006).

Thereversiblepost-translationalmodificationofproteinswith asingleubiquitinorwithubiquitinchainsisanimportantmech- anismtoregulatethefunction,activity,cellulardistributionbut alsothestabilityofproteinsbymediatingtheirproteolysisbythe 26Sproteasome.The ubiquitinproteasomesystem(UPS)is not onlyanimportantregulatorofproteindegradationbutalsoreg- ulates cancer-relevantprocesses such ascell cycle progression, proliferation,DNAdamageresponse,angiogenesisandapoptosis (Ciechanover,1998;HaglundandDikic,2005).Moreover,inhibitors oftheproteasomearealreadyinuseorunderfurtherinvestiga- tionforthedevelopmentofcancertherapies(ChenandDou,2010;

Tuet al.,2012).Ubiquitylation aswellaspoly-ubiquitylationis achievedbyanenzymaticcascadewherefirstubiquitinisadeny- latedatitsC-terminaldiglycineresiduebyoneofitstwoubiquitin E1activatingenzymesUBE1(Ciechanoveretal.,1981)orUBA6(Jin etal.,2007;Pelzeretal.,2007)andthentransferredontotheactive site-cysteineoftherespectiveE1enzymetoformathioesterbond.

Inasecondstep,ubiquitinistransferredtotheactivesitecysteineof acognateubiquitinconjugatingenzyme(E2)byatransthiolation reaction.Finally,differentclassesofubiquitinligases(E3s)catal- ysetheisopeptidelinkageofubiquitintothe␧-amino-groupofan internallysineresidueofasubstrateprotein(Finley,2009;Kerscher etal.,2006).Ubiquitinitselfcontains7lysineresidues(atpositions 6,11,27,29, 33,48and 63)whichareusedtobuildupchains ofdifferentlinkagetypesconveyingdifferentoutcomesforsuch modifiedproteins.Modificationofaproteinwithasingleubiqui- tinisnotenoughtoguideproteinstoproteasomaldegradationbut requirestheattachmentofaubiquitinchainofatleastfourubiqui- tinmolecules,linkedthroughoneoftheabovementionedinternal lysineresidues.Alldifferentlinkagetypes,withtheexceptionof K63,leadtothedegradationofthemodifiedproteinbythe26S proteasome(Xuetal.,2009),abarrelshapedmulti-enzymecom- plex,consistingofa20Scoreparticleandoneortwo19Sregulatory particles(Finley,2009).Beforedegradation,ubiquitinisremoved fromtheproteinsbysocalleddeubiquitylatingenzymes(DUBs) andrecycled(Reyes-Turcuetal.,2009;Wolberger,2014).

Besidesubiquitin,severalubiquitin-likemodifiers(ULMs)have beenidentified.TheULMsfamilyincludesHumanLeukocyteAnti- gen(HLA)-F adjacenttranscript10(FAT10),small-ubiquitin-like modifier(SUMO)-1/2/3,ISG15,NEDD8,ATG8,ATG12,UFM-1,and URM-1.TheULMsshareasimilarthree-dimensionalstructure,the ubiquitinor␤-graspfold,aswellastheC-terminaldiglycinemotif

foractivationandcovalentattachmenttosubstrateproteinsbyan E1–E3enzymaticcascade(Hochstrasser,2009).OfalltheseULMs, FAT10istheonlymodifierwhichactsasanautonomoustransfer- ablesignalfordegradationbythe26Sproteasomeanditwasshown thatthisprocesscanoccurindependentlyofubiquitin(Hippetal., 2005;Schmidtkeetal.,2009).FAT10isaproteinoftheimmunesys- temanditisstronglyup-regulatedbypro-inflammatorycytokines (Fan et al.,1996; Raasietal., 1999)and during thematuration of antigen presenting cells (Buerger et al., 2015;Ebstein et al., 2009;Lukasiaketal.,2008).Moreover,numerousrecentpublica- tionspointtoadirectinvolvementofFAT10incancerdevelopment.

Theserecentinsightswillbepresentedinthisreviewalongwith thecurrentstateofknowledgeontheregulationandfunctionof FAT10expressionininflammationandtumorigenesis(Fig.1).

2. Theubiquitin-likemodifierFAT10,aproteinofthe immunesystem

2.1. ThestructureofFAT10andhowittargetsforproteasomal degradation

TheFat10gene(originallydesignatedUbiquitinD(Ubd))was identifiedbygenomicsequencingofthehumanmajorhistocom- patibilitycomplex(MHC)byShermanWeissmanandcolleaguesin 1996(Fanetal.,1996).Thestructuralmodelofthe165aminoacid long,18kDaproteinFAT10aswellasapreviouslypublishedNMR structureoftheN-terminalubiquitin-likedomainofFAT10predicts twoubiquitin-likedomains,bothwithatypical␤-graspfold.TheN- andC-terminalubiquitin-likedomainsare29%and36%identicalto ubiquitin,respectively,arrangedinatandemarrayandseparated byashortlinker(Fanetal.,1996;Groettrupetal.,2008;Thengetal., 2014).Incontrasttoubiquitinandotherubiquitin-likemodifiers whichareexpressedasinactiveprecursorsandwhichneedtobe activatedbyproteolyticprocessingattheirC-terminusbyspecific proteases(Kerscheretal.,2006),FAT10isalreadysynthesizedasa matureproteinwithafreediglycinemotifatitsC-terminusandcan directlybeactivatedandconjugatedtosubstrateproteins(Raasi etal.,2001).AcentralfunctionofFAT10modificationistotarget proteinsfordegradationbytheproteasome(Hippetal.,2005;Raasi etal.,2001;Schmidtkeetal.,2014).Thisprocesswasshown to beindependentofubiquitylationbuttobedependentonanon- covalentinteraction partner of FAT10, namelyNEDD8-ultimate buster1long(NUB1L)(Hippetal.,2005;Schmidtkeetal.,2009, 2006).NUB1L,alikewiseinterferon-inducibleprotein(Kitoetal., 2001),actsasasolublereceptorandbringsFAT10andFAT10ylated proteinstotheproteasomewhereitbindstothe19Sregulatory subunitsRPN10 (S5a) andRPN1 (S2).It wasshown thatFAT10 itselfcouldalsodirectlybindtotheVWAdomainofRPN10inthe absenceofNUB1LbutthatthedockingofNUB1Ltotheproteasome wasnecessaryfortheaccelerateddegradationofFAT10(Ranietal., 2012;Schmidtkeetal.,2009,2006).Incontrasttoubiquitin,FAT10 hasaveryshorthalflifeofabout1handseemstobedegraded bytheproteasomealongwithitssubstrates(Aichemetal.,2014;

Hippetal.,2005).SinceNUB1Lnegativelyinfluencesthestability ofFAT10,theFAT10degradationratemightbedifferentindiffer- entcelllinesortissuesexpressingdifferentamountsofNUB1L,as e.g.inHEK293cells, whichexpressonlylowamountsofNUB1L andwhereFAT10ismorestable(Aichemetal.,2014).Inlinewith this,noFAT10-deconjugatingenzymes,whichmightcontributeto FAT10recycling,havebeenidentifiedtodate.

2.2. FAT10inadaptiveandinnateimmunity

TheFAT10geneisencodedintheMHCclassIlocusandthehigh- estFAT10mRNAexpressionismeasuredinorgansoftheimmune

(3)

Fig.1. SchemeshowingtheregulationofFAT10expressionaswellasdownstreamtargetswhichareinfluencedbyFAT10overexpressionintumortissues.

ThebroadandintenseoverexpressionofFAT10intumortissues,inducedbypro-inflammatorycytokinessuchasIFN-␥,TNF-␣andIL-6withinthetumormicroenvironment, contributestothemalignantcapacitiesofFAT10itself.FAT10regulatespathwaysinvolvedincancerdevelopmentsuchasNF-␬B-,Akt-,orWnt-signalingandalsodirectly interactswithandinfluencesdownstreamtargetssuchasp53,␤-catenin,SMAD2,andMAD2,leadingtoenhancedsurvival,proliferation,invasionandmetastasisformation ofcancercellsbutalsoofnon-malignantcells.Arrowsmarkedwitha“+”illustrateanactivatingpathway,arrowsmarkedwitha“−”illustratenegativelyregulatedpathways.

systemsuchasthymus,lymphnodes,fetalliverandspleen(Canaan etal.,2006;Leeetal.,2003;Liuetal.,1999;Lukasiaketal.,2008).

ThehighexpressionofFAT10inthethymuscouldbeallocated tomedullarythymic epithelialcells (mTECs)and it wasshown thatFAT10modifiesthymicT-cellselection,probablydue toan alteredpeptidepresentationonMHCclassImolecules(Buerger etal.,2015).AdditionaldatapointtoaroleofFAT10inMHCclassI antigenpresentation.Ebsteinandcolleagues(Ebsteinetal.,2012) showedthatFAT10couldenhancetheMHCclassIpresentation ofthehumancytomegalovirus(HCMV)-derivedantigenpp65in HeLacellsbyabouttwo-foldascomparedtountaggedpp65and Schlieheand colleagues(Schlieheet al.,2012)showedthatthe degradationrateaswellasthepresentationofspecificlymphocyte choriomeningitisvirus(LCMV)epitopesonMHCclassImolecules wasenhancedwhenFAT10wasfusedtotheN-terminusofthe LCMVnucleoprotein.ApartfromitsexpressioninmTECsFAT10is alsoconstitutivelyexpressedinmatureBcellsanddendriticcells (Batesetal.,1997)anditsexpressionishighlyandsynergistically inducibleonmRNAandproteinlevelinnumeroustissuesandcell typesbythepro-inflammatorycytokinesIFN␥andTNF␣(Aichem etal.,2010;Raasietal.,1999).Arecentpublicationshowed,that ahighinductionofFAT10expressionisalsoachievedbyasyner- gistictreatmentofHepG2cellswithTNF-␣incombinationwith IL-6(Choietal.,2014).UpregulationofFAT10expressioninduced caspase-dependentapoptosisinHeLacells,renaltubularepithe- lialcellsaswellasmousefibroblasts(Liuetal.,1999;Raasietal., 2001;Rossetal.,2006).However,theroleofFAT10inapoptosis iscontradictorybecauseotherpublicationsshowedapro-survival roleofFAT10(Canaanetal.,2006).LymphocytesofFAT10knock outmicewereshowntobemoresusceptibletospontaneouscell death than lymphocytes from wildtype mice and human and mousecardiacmyocyteswereprotectedfromapoptosisinduced bymyocardialinfarctionassociatedhypoxia/reoxygenationstress throughup-regulationofFAT10(Pengetal.,2013).

NexttotheroleofFAT10inadaptiveimmuneresponsesthere arerecentpublicationswhichassignafunctionininnateimmu- nitytoFAT10.FAT10wasshowntoplayaroleintheintracellular defense against bacteria (Spinnenhirn et al., 2014). Intracellu- larSalmonellaTyphimuriumwhich weretargetedforxenophagy

bybeingdecoratedwithubiquitin,theautophagyreceptorsp62 and NDP52as wellas withLC3weresimultaneouslydecorated withFAT10.Spinnenhirnandcolleagues(Spinnenhirnetal.,2014) showedthatthepresenceof FAT10resultedina prolongedlife span of S. Typhimurium-infected mice,pointing to a protective roleofFAT10.ItisassumedthatFAT10playsadditionalrolesin innateimmunitysinceanup-regulationofFAT10expressionwas described in HIV-infected renal tubular epithelial cells (RTECs) (Rossetal.,2006;Snyderetal.,2009)andduringKaposisarcoma- associatedherpesvirusinfection(Hongetal.,2004).Moreover,the groupofJ.-Y.YooshowedthatFAT10interferedwithRIG-I(retinoic acidinduciblegene1)mediatedantiviralsignalingnecessaryfor theproductionoftypeIinterferons.Bynon-covalentlyinteracting withRIG-I,FAT10sequesteredRIG-Iintoinsolubleaggregatesand thisresultedinadiminishedanti-viralimmuneresponse(Nguyen etal.,2016).

2.3. WhicharethetargetsofFAT10andhowisitconjugatedtoits substrates

UponIFN-␥/TNF-␣mediatedinductionofFAT10inthehuman embryonic kidney cell line HEK293, the FAT10 mRNA was detectable already after 4–5h, however, FAT10 protein was prominentlydetected inwestern blotsonly 24haftercytokine stimulation withthe highlysensitive monoclonalantibody 4F1 (Aichemetal.,2012,2010).Nevertheless,uponimmunoprecipita- tionofFAT10anditsmodifiedsubstrateswithmAb4F1,combined withasubsequentwesternblotanalysiswithapolyclonalFAT10 specificantibody,bulkFAT10conjugatesofalargerangeofsizesare detectable,similartoubiquitin,ISG15orSUMOconjugates.Amass spectrometryanalysisidentified 571putativeFAT10 interaction partners,ofwhich176proteinswereclassifiedasputativeconju- gationsubstratesandtheremaining395proteinsasnon-covalent interactionpartners.Interestingly,theseproteinsdidnotbelong todistinctcellularpathwaysbutcoveredalotofdifferentcellu- larfunctions,showingthatFAT10mightplayaroleinnumerous cellbiologicalprocesses(Aichemetal.,2012;Lengetal.,2014).Of importanceisthatFAT10apparentlyisnotonlyfunctionalwhen covalentlyconjugatedtoitssubstrateproteins,suchastheubiqui-

(4)

tinactivatingenzymeUBE1(Bialasetal.,2015;Ranietal.,2012), butalsowheninteractingnon-covalentlywithinteractionpartners suchasHDAC6(Kalverametal.,2008),aryl-hydrocarbonreceptor like-1(AIPL1),animportantproteinwithintheretina(Bettetal., 2012),ortheautophagosomalreceptorp62/SQSTM1whichinter- actsboth,covalentlyandnon-covalentlywithFAT10(Aichemetal., 2012).ActivationandconjugationofFAT10toitssubstrateproteins isperformedmostprobablybyathreestepenzymaticcascade,as describedforubiquitin.TheFAT10E1activatingenzymeUBA6acts asabi-specificE1enzyme,becauseitisabletoactivateboth,FAT10 andubiquitin(Chiuetal.,2007;Pelzeretal.,2007).Indoingso, UBA6isabletodistinguishbetweenthetwomodifiersandbinds FAT10withahigheraffinitythanubiquitin,althoughtheadeny- lationand transthiolationreaction isslowerfor FAT10than for ubiquitin(Gavinetal.,2012).TheactivatedFAT10isthentrans- ferredontotheactivesitecysteineofthelikewisebi-specificE2 conjugatingenzymeUBA6-specificE2enzyme1(USE1,alsocalled UBE2Z)whichdirectlyundergoesauto-FAT10ylationincis,mainly onLys323(Aichemetal.,2014,2012;Guetal.,2007;Jinetal.,2007).

Aportionoftheauto-FAT10ylatedUSE1issubsequentlydegraded bytheproteasome,albeit theremainingUSE1-FAT10conjugate remainsactivewithrespecttoFAT10andubiquitintransfer.There- fore,thismechanismmightrepresentanegativefeedbackloopto limittheFAT10conjugationpathway(Aichemetal.,2014).Sofarno FAT10-specificE3ligaseshavebeenidentified,butsinceallULMs whichgetcovalentlyconjugatedtosubstratesneedsuchligasesto befinallytransferredontotheirsubstrates,itisassumedthatsuch E3ligasesmustalsoexistforFAT10.

3. RegulationofFAT10expressionunderinflammatory conditionsandduringcancerdevelopment

3.1. FAT10expressionlevelsindifferentcancertypes

FAT10expressionishighlyup-regulatedinmanydifferentcan- certypessuchascoloncancer, hepatocellularcarcinoma(HCC), gastrointestinalandpancreaticcarcinomas,gynecologicalcarcino- mas,andglioma(Leeetal.,2003;Lukasiaketal.,2008;Qingetal., 2011;Sunetal.,2014;Yuanetal.,2012).InliverneoplasiaFAT10 isevendescribedasanepigeneticmarker(Frenchetal.,2010a,b;

Olivaetal., 2008).Moreover,inseveral cancertypesa statisti- callysignificantassociationbetweenahighFAT10expressionlevel andtheprogressionandseverityofthediseaseincludingahigher propensityformetastasisformationandpoorprognosis,hasbeen reported.Theseincludetriple-negativebreastcancer(Hanetal., 2015),glioma(Daietal.,2016;Yuanetal.,2012),pancreaticductal adenocarcinoma(Sunetal.,2014),HCC(Liuetal.,2014),orgastric cancer(Jietal.,2009).Furthermore,coloncancerpatientstreated withapostoperativechemotherapyandbearingtumorswithhigh FAT10expressiondisplayedahigherrecurrenceratethanthose withFAT10-negativetumorsandFAT10expressionwasassociated withasignificantlyshortersurvivaltime(Zhaoetal.,2015).

3.2. ThetransformingcapacityofFAT10

Lukasiaket al.(2008) showedthat thehighupregulation of FAT10expressionincancersofliverandcolonstronglycorrelated withtheexpression of the proteasomesubunit LMP2which is inducedintheseorgansbypro-inflammatorycytokinesIFN-␥and TNF-␣inthetumormicroenvironment.Byperformingacolonyfor- mationassaywithFAT10transfected NIH3T3cells,noevidence forcolonyformation andtransformation capacityofFAT10was observed,arguingagainstafunctionofFAT10asaproto-oncogene

inthisexperimentalmodel.Thereforeitwasquestionedwhether FAT10 hastransforming capacity and whether sucha property couldexplainitsoverexpressioninthesecancers.However,FAT10 overexpressionintheimmortalizedlivercelllineNeHepLxHTcould confermalignantpropertiestotheseotherwisenon-tumorigenic cells.NeHepLxHTcells,overexpressingclinicallyrelevantamounts ofFAT10,showedadose-dependenteffectofFAT10onprolifera- tion,anchorageindependentgrowth–ahallmarkoftransformed malignantcells–andresistancetoapoptosis(Gaoetal.,2014).In addition,ascompared tocontrolcells, FAT10expressingNeHe- pLxHTcellssignificantlyincreasedtheirinvasivecapacity across matrigelaswellastheirmigratorycapacityinascratch-wound- healingassay,againinaFAT10dose-dependentmanner.Thesame resultswereobtainedwhenusingasecondcelllinestablytrans- fectedwithaFAT10expressionplasmid,namelythetumorigenic colorectalcancercelllineHCT116.FAT10expressingNeHepLxHT cellsinducedtumorformation,againinadose-dependentmanner, wheninoculatedintonudemiceascomparedtomice,whichhad beentreatedwithemptyvector-transfected,FAT10-negativecon- trolcells.Furthermore,FAT10overexpressioninHCT116cellseven greatlyenhancedtumorgrowthascomparedtoFAT10-negative HCT116cells,stronglyarguingforadirectinvolvementofFAT10in tumorformation(Gaoetal.,2014).ThereforeitseemsthatFAT10 cannotonlyconfermalignantpropertiestonon-tumorigeniccells butthatitcanevenaugmentthemalignancyassociatedtraitsofa cancercellline.

AdditionaldataalsosuggestedFAT10tobeaproto-oncogene:

knockdownofFAT10intheFAT10-positivehepatocellularcarci- nomacelllineHep3Bspecificallyinhibitedcellproliferationand colony formation under in vitroconditions, most probably due toablockageoftheentryintotheS-phaseofthecellcycleand byinducingapoptosis,asmeasuredbyAnnexinVstaining.Under invivoconditionsusingnudemicewhichhadreceivedasubcuta- neousinjectionofHep3BcellsitwasfurthershownthattheHep3B xenograftgrowthwasmuchreducedwhenthemiceweretreated withanintratumoralinjectionofAd-siRNA/FAT10twoweeksafter injectionoftheHep3Bcells.TheknockdownofFAT10evenpro- longedthelifespanofthesemiceascomparedtomice,whichhad beentreatedwithacontrolvirusorPBS(Chenetal.,2014).

ThenotionthatFAT10isdirectlyinvolvedinthedevelopment oftumorsisatleastconsistentwithrecentinvestigationsofFAT10 geneknockoutmice.Thesemiceareviableandfertileandexhibit onlyminimalphenotypicdifferencestotheirwildtypecounter- partssuchase.gahighersensitivitytowardsendotoxinchallenge.

Inaddition,asalreadymentioned,lymphocytesfromthesemice aremorepronetospontaneousapoptoticcelldeath(Canaanetal., 2006).Noteworthy,whenexaminingolderFAT10-deficientmice, itcouldbeobservedthatthesemiceshoweddelayedagingand anappox.20%increaseinlifespan.Thisphenotypewasaccompa- niedwithasignificantreductioninadiposetissueandanincreased metabolisminmuscletissue.InmusclesofFAT10deficientmice, decreasedlevelsofpro-inflammatorycytokinesandenhancedlev- elsofanti-inflammatorycytokinessuchasIL-10weremeasured, therebycreatinganinflammation-suppressivemilieuinskeletal muscleandbloodwhichmayaccountfortheabsenceoftumorsin thesemice(Canaanetal.,2014).

Taken all these findings together, several publications sup- port thenotion that FAT10itself is a proto-oncogene and that itsupregulationduringinflammatoryprocessesisinvolvedinthe developmentofdifferentcancertypes.Thisconceptionposesfur- therquestions:(1)HowistheexpressionofFAT10regulatedduring inflammationandcancer?(2)Whicharethedownstreamtargetsof FAT10?and(3)HowdoesFAT10enhancethemalignantproperties oftheseproteins.

(5)

4. WhatarethemechanismsbehindFAT10overexpression?

What are the molecular mechanisms how FAT10 exerts its malignanteffects?Onepotentialmechanismcouldbebasedona recentlydescribedinvolvementofFAT10inNF-␬Bsignaling(Fig.1).

NF-␬Bis animportanttranscription factorfor theregulationof genesinvolvedininflammation,proliferation,cell-cyclecontrol, apoptosis,andtheimmuneresponseandplaysthereforeacriti- calroleincarcinogenesis(HoeselandSchmid,2013).Ontheone handFAT10mayactasaregulatorofNF-␬Bsignalingandonthe otherhandFAT10expressionitselfisregulatedbyNF-␬B.Asalready pointedout, FAT10mRNAand proteinexpressionis highlyand synergisticallyinducibleinallcelltypesbythepro-inflammatory cytokinesIFN-␥andTNF-␣(Aichemetal.,2010;Raasietal.,1999;

Renetal.,2011)orbyacombinationofTNF-␣andIL-6asshown inHepG2cells(Choietal.,2014).InHCT116cellsitwasshown, thatTNF-␣signalingviatheTNF-␣receptor1 (TNFR1)induced FAT10expression,areceptormainlyfoundontumorandstromal cellswhereitmediatestheactivationofNF-␬B(Balkwill,2009;Ren etal.,2011).TheanalysisoftheFAT10promotersequencerevealed sevenpotentialp65/NF-␬Bbindingsitesaswellasthreebinding sitesfortranscriptionfactorsofthefamilyofsignaltransducerand activatoroftranscription(STAT)3(Canaanetal.,2006;Choietal., 2014;Gaoetal.,2015).NF-␬BandSTAT3cooperativelyregulate anumberoftargetgenesanditwasshownthatbindingofone ofthesetranscriptionfactorstotherespectivepromotersequence facilitatesrecruitmentoftheother,andviceversa.STAT3activa- tionismediatedmainlybyIL-6andthisleadstotheexpression ofgenesinvolvedincellproliferation,angiogenesisandsurvival.

STAT3isconstitutivelyactiveinmanycancertypessuchasbreast andcoloncanceranditsexpressionlevelpositivelycorrelateswith theaggressivenessof thetumor (Bosch-Barrera andMenendez, 2015;HoeselandSchmid,2013;WakeandWatson,2015).Choi etal.(2014)haveshownthatSTAT3andp65/NF-␬Bactsynergisti- callytoactivateFAT10expressionuponIL-6andTNF-␣induction.

FurthersupportfortheregulationofFAT10expressionbyNF-␬B signalingisderivedfromexperimentswithasubstancecalledsilib- inin,aflavonoidisolatedfromseedextractsoftheherbmilkthistle (Silybummarianum).Silibininexhibitsseveralpotentanti-cancer propertiessuchasactingasanantioxidant,havingimmunomodu- latoryandanti-proliferativepropertiesanditwasshowntoinhibit multiplecancersignalingpathwaysasforinstanceNF-␬Bsignal- ing(Bosch-BarreraandMenendez,2015;Singhetal.,2004;Tyagi etal.,2012).CarolineLeeandcolleaguesfoundthatupontreatment ofIFN-␥/TNF-␣-stimulatedHCT116cellswithincreasingamounts of silibinin, the FAT10 mRNA expression was diminished in a dose-dependentmannerandthiswasduetoasilibinin-mediated preventionofthenucleartranslocationofp65mostprobablydue toaninhibitionofthedegradationoftheNF-␬BinhibitorI␬B␣(Gao etal.,2015).TumorformationbyTNF-␣-inducedandthusFAT10 expressingHCT116cellsinnudemicewasdiminishedwhenthe miceweretreatedatthesametimewithsilibininwhichcorrelated withreducedFAT10expressioninthetumors.Thedependencyof endogenousFAT10expressiononNF-␬Bsignalingwasfurthersup- portedbydatashowingthattheinductionofFAT10expressionby TNF-␣wasgreatlydiminishedwhenp65wasdownregulatedby specificsiRNAsinHCT116andHepG2cells(Gaoetal.,2015;Ren etal.,2011).

TheinterplaybetweenFAT10and NF-␬Bmaynotbea one- waystreetsinceinapreviousstudyusingrenaltubularepithelial cells, FAT10was reportedtoactivate NF-␬B signaling,pointing to a potential positive feedback loop. Consistently, the TNF-

␣-induced I␬B␣ degradation, as well as the expression of the NF-␬B-dependentgenesCXCL2andMCP-1wasimpairedinFAT10- deficient cells (Gong et al., 2010).Arguing against a RTEC cell type-confined effectofFAT10 onNF-␬Bsignaling,a microarray

basedtranscriptomeanalysisofFAT10overexpressingNeHepLxHT andHCT116cellsrevealedinbothcelllinesthattheNF-␬Btran- scriptional activity was significantly enhanced as compared to controlcells, notexpressingFAT10.Thiswasaccompaniedbya highernuclearlocalizationofNF-␬B/p65,whilethetotalcellular amountofp65remainedstable.Atthesametime,theamountof theNF-␬BinhibitorI␬B␣wasdiminishedinFAT10overexpressing NeHepLxHTcells.Asimilarincreaseinnuclearp65wasobserved inHCT116cells,overexpressingFAT10.Theincreaseinnuclearp65 couldbereversedbyknockingdownFAT10withFAT10-specific siRNAs(Gaoetal.,2014).Takentogether,itwasshownthatthe increasedFAT10expressionindirectlyaugmentedNF-␬Bactivity byalleviatingtheinhibitionofNF-␬B.NF-␬Bisatranscriptionfac- torwithmultipletargetgenes,sothequestionarises,whicharethe genesdownstreamofFAT10andNF-␬B?Themicroarrayexpression analysisofFAT10overexpressingNeHepLxHTandHCT116cellsby Gao etal.(2014)revealedthat thechemokinereceptorsCXCR4 and CXCR7 were highly up-regulated in FAT10 overexpressing NeHepLxHTcells,consistentwithotherdatashowingthatNF-␬B isa transcriptionfactorforboth genes(KatohandKatoh,2010;

Tarnowskietal.,2010).CXCR4and−7arebothdescribedtopro- moteinvasionandtumorgrowthinseveralcancertypes(Gaoetal., 2010;Heinrichetal.,2012;Liangetal.,2005;Zhengetal.,2010).In accordancewiththesedata,ansiRNA-mediatedknockdownofp65 oroverexpression ofI␬B␣inFAT10overexpressingNeHepLxHT cellssignificantlyattenuatedcellinvasionandmigrationinvitro, precisely assilencingofCXCR4 andCXCR7 did.Taken together, these datapoint towards a mechanism where up-regulationof FAT10leadstoactivationofNF-␬Bsignalingwhichinturnactivates theexpressionofCXCR4and−7,resultinginenhancedmigration andinvasionofFAT10overexpressingcells(Gaoetal.,2014).

Insummary,itemergesthatFAT10expressionuponTNF-␣/IFN-

␥ or TNF-␣/IL-6 stimulation is mediated mainly by NF-␬Band STAT3signaling.Thereby,a FAT10-dependent positivefeedback loopseemstoexistwhichenhancesNF-␬Bactivationandwhich maycontributetothemalignantpropertiesofFAT10.Thisdirectly leadstothenextquestion,namelywhichproteinsgettargetedby FAT10and whataretheconsequences oftheirinteractionwith FAT10withrespecttocancerdevelopment.

5. FunctionalconsequencesofFAT10up-regulationin inflammationandcancer

5.1. MAD2andchromosomalinstability

UpregulationofendogenousFAT10expressionbyTNF-␣aswell asoverexpressionofFAT10hasdirectlybeenlinkedtochromoso- malinstabilityinseveralreports(Gaoetal.,2015;Renetal.,2006, 2011;Thengetal.,2014).Itwasshownthatabout60%ofHCT116 andHepG2cellswhichhadbeentreatedforabouttenpassageswith IFN-␥andTNF-␣displayedabnormallyhighchromosomenumbers inakaryotypeanalysisandinhibition ofFAT10expressionwith silibinin,apotentinhibitoroftheNF-␬Bsignalingpathway,was abletoabolishthiseffect(Gaoetal.,2015).FAT10wasshownto becell-cycleregulatedwithapeakofexpressioninS-phasebut low expressionlevelsduring G2/Mphase (Lim etal.,2006; Liu etal.,2014)andaproteomicsbasedstudyprovidedevidencefor aroleofFAT10inmitoticregulation(Merbletal.,2013).Already 17yearsagoMAD2(Mitoticarrestdeficient2),amitoticspindle checkpointprotein,wasidentifiedasanon-covalentinteraction partnerofFAT10inayeasttwohybridscreenusingahumanBcell cDNAlibrary(Liuetal.,1999)andthisinteractionwasshownto prevailduringmitosis(Renetal.,2006).MAD2bindstothekine- tochoresofsisterchromatidsinthemetaphaseofthecellcycle.

Thereit ensures thatbeforetheonset of anaphaseallchromo-

(6)

Fig.2.InteractionofFAT10withMAD2inducesaprematureonsetofanaphase,resultinginaneuploidy.

(A)Undernormalconditions,theonsetofanaphaseandtheseparationofthesisterchromatidsisinhibiteduntilallchromosomesareproperlyattachedviatheirkinetochores tothemitoticspindleapparatus.ThismechanismissurveilledbyaproteincomplexcontainingMAD2.(B)UpregulationofFAT10allowsthenon-covalentinteractionof FAT10withfreeMAD2andinturnreducestheamountofMAD2atthekinetochores.Asaresult,anaphasestartstooearlyandresultsininappropriateseparationofsister chromatidsandaneuploidy.Adetaileddescriptionisgiveninthetext.

somesbecomeproperlyattachedtothemitoticspindlealongthe metaphaseplate(Fig.2A).MAD2achievesthisbyinteractingwith andinhibitingCdc20,aco-activatorandsubstraterecruitmentfac- toroftheanaphase-promotingcomplex/cyclosome(APC/C),which isamulti-subunitubiquitinE3ligase.Therebytheubiquitylation ofsecurinandcyclinisprevented.Securinbindsandinhibitsapro- teasecalledsecurasewhichcleavescohesin,aproteinthatkeeps thetwo sister chromatidstogether (Lara-Gonzalezet al., 2012;

MusacchioandSalmon,2007).InFAT10overexpressingHCT116 cellsaswellasinTNF-␣-treatedHCT116cells,thelocalizationof MAD2atthekinetochoreswasmuchreducedintheprometaphase ofthecellcycle,indicatingthatFAT10interfereswiththebindingof MAD2tokinetochores(Fig.2B)(Renetal.,2006,2011).Moreover, afterreleasefromG1/Sarrest,FAT10overexpressingcellsshowed a delayedentryintomitosis. However,thesecells alsoshowed anunalteredreentryintoG1phaseascomparedtoparentalcells whichpointsatanabbreviatedmitoticphaseoranabbreviated mitoticarrestuponspindledamage.Whencomparedwithcontrol cells,ahighernumberofFAT10overexpressingcellsfailedtoarrest inmitosisupontreatmentwithnocadozoleorevenescapedthe mitoticarrest.Concurrentwiththesefindings,FAT10overexpres- sioninducedtheformationofmultinucleatedcellswithabnormal nuclearmorphology.A constitutive overexpression of FAT10as wellasconstitutivetreatmentwithTNF-␣resultedinanincreased chromosomenumberascomparedtotheparentalHCT116cellline (Renetal.,2006,2011).However,whenthefiveputativeinterac- tionsitesofMAD2withintheN-terminalubiquitin-likedomainof FAT10weremutated(H11D,R13Q,H75D,T77DandK79Q),bind- ingofMAD2toFAT10wasgreatlydiminished.Ascomparedto cellsoverexpressingwildtypeFAT10,thechromosomenumbersof cellsexpressingthemutantFAT10returnedtonormalasfound inwildtypeHCT116cells.Thisfurthersupportsthenotionthat FAT10overexpressioncauseschromosomalinstabilitybyinteract- ingwithMAD2.Moreover,ascomparedtotheparentalcells,FAT10 overexpressingcellsshowedlong,incompletelycondensedchro- mosomesandthiswasalsorevertedbacktonormallycondensed, short“ribbon-like”chromosomes incellsexpressingthemutant

FAT10deficientinMAD2binding.Inlinewiththisobservation,the inhibitionoftheFAT10andMAD2interactionalsorevertedthe capabilityofFAT10overexpressingcellstoescapethenocadozole- inducedmitoticarrestbacktolevels,seenwithwildtypeHCT116 cells(Thengetal.,2014).Takentogether,thesedatacorroboratethe hypothesisthatthenon-covalentinteractionofFAT10andMAD2 is critical for thedevelopment of aneuploidyin cancer cells. It wasshownthat FAT10interacts withfree butnot withspindle boundMAD2asanimmunoprecipitationagainstFAT10ledtoaco- immunoprecipitationofMAD2only,andnotofadditionalproteins ofthespindlecomplexsuchasMAD1andBub1(Thengetal.,2014).

TheobservedlowlevelofendogenousFAT10expressioninG2/M phase(Limetal.,2006)suggeststhatcellskeeptheFAT10level lowduringthemitoticphaseinordertopreventmitoticdysfunc- tionswiththeriskofsubsequentcancerdevelopment.Consistently, HCT116cellsoverexpressingtheMAD2interaction-deficientFAT10 mutantretainedsignificantlylowerproliferationratesandshowed anincreasedsusceptibilitytoapoptosisascomparedtocellsover- expressingwildtypeFAT10.Moreover,cellsexpressingtheMAD2 binding-deficientFAT10mutant failedtogrowin soft agarand losttheircapabilitytogrowinananchorageindependentman- ner.Moreimportantly,thisfindingcouldbesubstantiatedinvivo byinjectingHCT116cellsexpressingFAT10wildtypeortheMAD2 binding-deficientFAT10mutant intonudemice.Tumorsgrown fromHCT116cellsexpressingMAD2binding-deficientFAT10were significantlysmallerthanthoseexpressingwildtypeFAT10(Theng etal.,2014).Insummary,allthesedatafromwellcharacterizedcell linemodelsofhepatocellularcarcinomamakeaconvincingcase, thatthepro-malignantcapacitiesofFAT10relyextensivelyonits interactionwiththespindlecheckpointproteinMAD2,therebypre- ventingtheproperfunctionofMAD2inmitoticcheckpointcontrol (Fig.2).

5.2. p53

Thetranscriptionfactorandtumorsuppressorp53playsacen- tralroleinthemaintenanceofgenomicstabilityandasaninhibitor

(7)

Fig.3. FAT10negativelyregulatesp53transcriptionalactivity.

Undernormalconditions,theconcentrationofp53iskeptatalowlevelandthisismaintainedbyatightregulationofp53ubiquitylationanddeubiquitylationbyspecific E3ligasesanddeubiquitylationenzymes(DUBs).DNAdamageleadstoarapidupregulationofp53,itsactivationbyphosphorylationanditstrans-localizationintothe nucleuswhereitactseitherasanactivatororasaninhibitoroftranscriptionleadingtoDNArepairorinductionofapoptosis.HighFAT10concentrations,mediatedby pro-inflammatorycytokinesleadtoaninhibitionofthep53transcriptionalactivityandresultinDNAdamageandresistancetoapoptosis.Adetaileddescriptionisgiven inthetext.Arrowsmarkedwitha“+”illustrateactivatingpathways,arrowsmarkedwitha“−”illustratenegativelyregulatedpathways.Aredarrowwithtwoarrowheads representstheinteractionofp53andFAT10.

ofinflammation.Morethan50%ofallhumancancersdisplaydele- tionsormutationsinthep53gene.Undernormalconditions,the concentrationofp53iskeptatalowlevelandthisismaintained byatightregulationofp53ubiquitylationanddeubiquitylation.

Twomaincellulareventsregulatedbyp53arecell cyclearrest uponDNAdamageandinductionofapoptosis.DNAdamageleads toarapidup-regulationofp53,itsactivationbyphosphorylation anditstrans-localizationintothenucleuswhereitactseitheras anactivatororasaninhibitoroftranscription.Down-streamtar- getsof p53aregenesinvolved intheregulationof DNArepair, apoptosisandcellcyclecontrol.Uponup-regulationofp53,the cellcycleisarrestedatthetransitionofG1/SuntilDNArepairis performedandthecellre-entersthecellcycle.However,ifthe DNAdamageistoostrong,apoptosiswillbeinduced.Mutationsin p53whichinhibititsactivityastranscriptionfactoraretherefore linkedtocancerdevelopment(Gudkovetal.,2011;Levine,1997).

p53wasrecentlyidentified as aFAT10 interactingproteinin a massspectrometryscreensearchingforFAT10interactionpartners (Aichemetal.,2012).Additionaldatashowedthatp53getscova- lentlymodifiedbyoverexpressedFAT10inHEK293cells,andthat overexpressionofwildtypebutnotofmutantFAT10,inwhichthe diglycinemotifattheC-terminuswasmutated,increasedthetran- scriptionalactivityofa luciferasereporterconstruct,containing bindingsidesforp53(Lietal.,2011).However,underinflamma- toryconditionsthep53transcriptionalactivitytowardsluciferase reporter constructs containingeitheran artificialp53-regulated promoteroranaturaltargetpromoterofp53(p21WAF1/CIP1)was decreased upon FAT10induction byIL-6 and/orTNF-␣.Consis- tently,overexpressionofFAT10indifferentcellslinesalsoreduced thetranscriptionalactivity ofp53 withoutaffecting theoverall

p53proteinamount.Insupportofthisfinding,silencingofFAT10 induced theopposite effectand ledtoan increase inthe tran- scriptionalactivityofp53andtoanincreaseinapoptosis(Choi etal.,2014).Thesystemseemstobetightlyregulatedbyaneg- ativefeedbackmechanismbecauseitwasshown,thatp53itself negativelyregulatestheexpressionofFat10mRNAbybindingto aninhibitorysitewithintheFat10promoter.Itwasshownthat thetranscriptionalactivityof␤-galactosidasereporterconstructs underthecontroloftheFat10promoterwassignificantlyhigherin thep53negativecelllineHep3Bascomparedtothep53expressing celllinesHepG2andKB3-1.Overexpressionofp53inHep3Blike- wisedecreasedFat10reporteractivityaswellasendogenousFAT10 expression.Conversely,whenp53wasdownregulatedbyspecific siRNAinHep3Bcellsoverexpressingp53,Fat10reporteractivity increased.ThesameincreaseinendogenousFAT10mRNAexpres- sionwasobtainedwhenendogenousp53wasdownregulatedby specificsiRNAinHEK293cells,clearlyunderliningthatp53nega- tivelyregulatestheFat10promoter.Thesedataimplythatmutant p53,asfoundinmanytumors,wouldelevateFAT10expressionand alongwithitFAT10‘spro-malignantcapacities(Zhangetal.,2006).

Insupportofthishypothesis,themRNAandproteinexpression levelsofFAT10positivelycorrelatedwiththemRNAandprotein expressionlevelsofmutantp53ingastriccancertissueandpatients withhighFAT10expressionintheirtumorsshowedatendency towardsunfavorableprognosisregardingtheoverallsurvivalrate (Jietal.,2009).

Taken allthese datatogether,one canconstruct amodel of howtheexpressionlevelsofp53andFAT10mustbetightlyregu- latedtopreventcellsfrombecomingmalignant(Fig.3).Innormal cellsexpressingfunctionalp53,FAT10expressioniskeptatalow

(8)

Fig.4.FAT10regulates␤-cateninstabilitybyinfluencingtheWntandAktsignaltransductionpathways.

TheWntsignalingpathwayplaysacriticalroleinpromotingHCCcarcinogenesisandmetastasis.Itleadstotheactivationofthetranscriptionfactor␤-cateninbypreventing itsphosphorylationbyGSK3␤andthusitssubsequentubiquitylationandproteasomaldegradation.FAT10contributestothemalignantcapacitiesof␤-cateninbydirectly interactingwithitandinhibitingitsubiquitin-dependentdegradationbytheproteasome.Inasecondline,upregulationofFAT10leadstoanincreaseofphosphorylatedand thusactivatedAkt,whichinturnphosphorylatesandinactivatesGSK3␤,whichagainresultsinstabilizationof␤-catenin.Adetaileddescriptionisgiveninthetext.Anarrow markedwitha“+”illustratesanactivatingpathway,anarrowmarkedwitha“−”illustratesanegativelyregulatedpathway.Redarrowwithtwoarrowheadsrepresentsthe interactionof␤-cateninandFAT10.

levelduetothenegativeregulationoftheFAT10promoterbyp53.

IncaseofDNAdamage,cellcyclearrestandDNArepair isper- formedtoletthecellre-enterthecellcycle,or,iftheDNAdamage istoosevere,apoptosisisinduced.Chronicinflammatorycondi- tionswhichinducehighFAT10expressionleadtoatranscriptional downregulationofp53-dependentgeneexpressionandpromote theinteractionofFAT10withMAD2.Thisleadstoatooearlyonset ofanaphase, chromosomal instabilityand protectionfromapo- ptosis.Thepresence ofmutant, non-functional p53might even enhancethiseffectandfurthersupportthetransformingcapaci- tiesofFAT10.ThemaintenanceofthebalancebetweenFAT10and p53expressionisthereforeveryimportanttoprotectcellsfrom gettingmalignant.

5.3. ˇ-catenin

Thewntsignalingpathwayleadingtotheactivationofthetran- scriptionfactor␤-cateninplaysacrucialroleinthedevelopmentof cancer.␤-catenin,aslongasitisnotphosphorylated,cantranslo- cateinto the nucleus where it forms heterodimers with other transcriptionfactorssuchasT-cellfactorandlymphoidenhancer- bindingfactor1,leading tothetranscriptionof genes,involved intheregulationofproliferation,differentiationandcellsurvival (Fig.4).Uponphosphorylationof␤-cateninbytheserine/threonine glycogensynthasekinase3␤(GSK3␤)andbycaseinkinaseI,␤- cateningetsubiquitylatedbytheE3ligase␤-TRCPanddegradedby theproteasome(Amitetal.,2002;Polakis,2000,2012).Mutations in␤-cateninwhichpreventitsphosphorylationandsubsequent ubiquitylation,andwhichresultinaconstitutiveactivetranscrip- tion factor are found withsignificant frequency in HCC and it wasshown that␤-cateninplays akeyrole inthemetastasisof humanhepatocellularcarcinoma(deLaCosteetal.,1998;Laietal., 2011).Recently,itwasreportedthattheup-regulationofFAT10 expressionpromotedtheinvasion andmetastasisof hepatocel- lularcarcinomacellsbybindingnon-covalentlyto␤-cateninand preventingitsubiquitylationandsubsequentproteasomaldegra- dation(Fig.4).Asaresult,theexpressionofHomeoboxB9(HOXB9),

amember oftheclassIhomeobox(HOX)genesand atargetof

␤-catenin/TCF4(Hatzisetal.,2008),wasfoundtobehighlyup- regulatedin HCCandtheHOXB9mRNAandproteinexpression levelscorrelatedwiththemRNAandproteinexpressionlevelsof FAT10.HOX genesarenormally involved inembryogenesis but a role in cancerdevelopment has been proposed(Abate-Shen, 2002).HOXB9regulatestheexpressionofgenesinvolvedinthe regulationof cellmigration, invasionand metastasisformation.

FAT10andHOXB9overexpressioncorrelatedwithHCCtumorsize, tumormicrosatelliteformationandvascularinvasion,pointingto an involvement of both proteinsin HCC aggressiveness. Taken together,thesedatashowthatFAT10up-regulationstabilizesthe

␤-catenin/TCF4 signaling pathway by preventing its ubiquitin- dependentproteasomaldegradation,andthisinturninducesan increaseinHOXB9expression,leadingtoHCCinvasionandmetas- tasisgeneration(Yuanetal.,2014).

Concurrentwiththesefindings,FAT10wasfoundtoinfluence theAkt/GSK3␤signalingpathway(Fig.4).The serine/threonine kinaseAkt(orPKB)isacentralnodeincellsignalingdownstream ofcytokines,growthfactorsandotherstimuli.Aktsignalingplays aroleinregulatingcellsurvival,differentiation,proliferationand angiogenesis,pointing toa roleoftheAktsignalingpathwayin carcinogenesis(Manningand Cantley, 2007).One ofthedown- streamtargetsofAktisGSK3␤,whichisinactivatedwhengetting phosphorylatedbyAkt,andsignaltransductionviaAkt/GSK3␤was showntopromoteTNF-␣-mediatedepithelial-mesenchymaltran- sition(EMT)(Wangetal.,2013).FAT10overexpressingHCCcell linesHepG2andSMMC-7721wereshowntoundergoEMTbecause theirmorphologychangedtowardsspindle-likefibroblasticcells, theexpressionoftheepithelialmarkerE-cadherinwasdecreased, andtheexpressionofthemesenchymalmarkerN-cadherinand vimentinwereincreased.Inaddition,theamountofsnail,anE- cadherin transcriptional repressor, was elevated in these cells.

FurtherinvestigationsshowedthatFAT10mediatedthiseffectvia theAkt/GSK3␤pathway.PhosphorylatedandtherebyactivatedAkt wasfoundtobehighly elevatedinFAT10overexpressing cells,

(9)

accompaniedbyanincreaseintheamountofphosphorylatedand thusinactivatedGSK3␤andastabilizationof␤-catenin(Liuetal., 2014).Anincreasein␤-catenininHCCwasalreadyreportedear- lierandit hadbeenshownthat thiscontributestodegradation of E-cadherin and inductionof EMT(Nejak-Bowen and Monga, 2011).In summary, FAT10seemsto elicitits stabilizing effects onto␤-cateninleadingtocellmigration,invasionandmetastasis ofHCCintwoways(Fig.4).Inafirstline,FAT10directlyinteracts with␤-cateninwhich prevents itsubiquitylation andproteaso- maldegradationleading toincreased␤-catenin levelsand thus toincreasedexpressionof downstream targetssuchasHOXB9.

Inasecondline,overexpressionofFAT10elevatestheamountof activeAkt,whichinturnphosphorylatesGSK3␤,renderingitinac- tivewhichinturnstabilizes␤-catenin.TheAkt-GSK3␤signaling pathwayisthereforeanotherintriguingmechanismofhowFAT10 exertsitscarcinogeniceffectsinlivercarcinoma.

5.4. Smad2

Still another downstream target of FAT10 wasidentified by investigating the contribution of FAT10 upregulation onto the developmentofglioma,aneuroepithelialtumorarisingfromglia cellsandaccountingfornearlyhalfofallintracranialtumors(Filbin andSuva,2016).Gliomasareclassifiedintofourgrades(I–IV)with increasingseverity.FAT10isprominentlyexpressedin gliomas, withthehighestmRNAandproteinexpressioningradeIVgliomas.

TheincreaseinFAT10expressionpositivelycorrelateswithpro- gressionofthediseaseandFAT10-positivepatientshavethelowest survivalrate(Daietal.,2016;Yuanetal.,2012).Gliomacelllines stably overexpressingFAT10 displayedenhanced tumorigenesis invitrobecausetheydevelopedsignificantlymorespheresandpos- sessedincreasingmigrationandinvasioncapacity.Wheninjected intonudemice,thesecellsdevelopedlargertumorsascompared tomock-transfectedcells,implicatingthatFAT10isalsopromoting gliomacarcinogenesis.FAT10seemstoexertitseffectonglioma- genesisviathetransforminggrowthfactorbeta(TGF␤1)signaling pathway.WhereasinnormalcellsTGF␤inhibitsproliferation,it turnsouttobeanoncogenicfactorinhighgradegliomabecause itinfluencesgliomaproliferation,survival,migration,invasionand theescapefromhostimmunity.DimericTGF␤bindingtooneofthe TGF␤typeIandIIreceptordimersinducesconformationalchanges andactivateskinasedomainsofthereceptors.Thisleadstothe phosphorylationandactivationofSmad2andSmad3transcription factors,whichinteractwithSmad4andaccumulateinthenucleus wheretheyregulatethetranscriptionofTGF␤expressionitselfbut alsoofgenese.g.involvedincellcycleregulation(Kaminskaetal., 2013).OverexpressionofFAT10inthegliomacelllineU251ledto significantlyincreasedlevelsofphosphorylatedSmad2(p-Smad2) andofothercancerstem-cellmarkerssuchasCD133,OCT4,nestin, andBmi-1.Inturn,knockdownofFAT10ortheapplicationofan inhibitoroftheTGF␤signalingpathway(LY364947)reversedthis effect.Althoughtheexactmechanism ofthis functionofFAT10 needsfurtherclarification,itappearsthataberrantFAT10might influencetheprogressionofgliomabyenhancingthelevelofp- Smad2(Daietal.,2016).

6. Summaryandfutureprospects

Growing evidence speaks for a direct involvement of the ubiquitin-like modifier FAT10 in tumorigenesis. Recent data strongly suggests that FAT10itself promotescarcinogenesis by directlyincreasingsurvival,proliferation,invasion,andmetastasis formationoftumorcellsassummarizedinFig.1.GiventhatFAT10 isapotentsignalforproteasomaldegradation,itisremarkablethat otherthantheFAT10ylationofp53nocovalentsubstratesofFAT10

conjugationwithpotentialfortumorpromotion(e.g.tumorsup- pressorsorapoptosisinducers)havebeendescribed.SinceFAT10 getsisopeptidelinkedtohundredsofproteins,thisisclearlyafield for future investigations.Anothercentralopenquestion iswhy FAT10,whenectopicallyoverexpressedmainlyinnontransformed cells,potently inducesapoptosiswhileitseemstopromotecell growthandsurvivalinothercells,inparticularincancerderived celllines.ThestrongincreaseinFAT10expressionin anumber ofdifferenttumortypesmightbeusefulasaprognosticmarker for thesurvival rateoftheaffectedpatientsand forestimating theprobabilityofrecurrence.Potentialtherapeuticavenuesaim- ingatFAT10-inducingpathwayssuchasNF-␬B/STAT3signaling aswellastargetingtheFAT10conjugationpathwayseemtohold promiseinparticularbecausethegeneticablationofFAT10inmice doesnotcausesevere phenotypes.Apossiblestarting pointfor targetingtheFAT10conjugationpathwaymightbeaninhibitor of the FAT10 E1 activating enzyme UBA6, developed by Gavin andcolleagues,whichwasalreadysuccessfully testedtoinhibit ubiquitinactivationbyUBA6(Gavinetal.,2012).However,since UBA6isabispecificE1activatingenzymeforFAT10andubiqui- tin,thisoptionmustbehandledwithcaretominimizeputative sideeffects.Targetingtheubiquitin-proteasomesystemincanceris alreadyclinicallyeffectiveatleastviainhibitionoftheproteasome whileinhibitorsoractivatorsofE3ligasesorDUBswithabetter perspectiveforselectivityarebeingdeveloped(Weathingtonand Mallampalli,2014).ThesearchforFAT10-specificE3ligasesaswell asforFAT10-specificdeconjugatingenzymesisstillongoingand theidentificationoftheseenzymeswillenableinvestigationson whethertargetingoftheseenzymeswillprovidenewoptionsfor thedevelopmentoffuturepharmaceuticalsagainstcancer.

Conflictofinterest

Theauthorsdeclarenoconflictofinterest.

Acknowledgements

ThisworkwasfundedbygrantsfromtheCollaborativeResearch Center969,ProjectC01,oftheGermanResearchFoundationand bygrantNrs.855and1029fromtheVeluxfoundation.Thefund- ingsourceshadnoinfluenceonwritingofthisarticleandonthe publicationdecision.

References

Abate-Shen,C.,2002.Deregulatedhomeoboxgeneexpressionincancer:causeor consequence?Nat.Rev.Cancer2(10),777–785.

Aichem,A.,Pelzer,C.,Lukasiak,S.,Kalveram,B.,Sheppard,P.W.,Rani,N., Schmidtke,G.,Groettrup,M.,2010.USE1isabispecificconjugatingenzymefor ubiquitinandFAT10,whichFAT10ylatesitselfincis.Nat.Commun.1,13.

Aichem,A.,Kalveram,B.,Spinnenhirn,V.,Kluge,K.,Catone,N.,Johansen,T., Groettrup,M.,2012.TheproteomicanalysisofendogenousFAT10substrates identifiesp62/SQSTM1asasubstrateofFAT10ylation.J.CellSci.125, 4576–4585.

Aichem,A.,Catone,N.,Groettrup,M.,2014.Investigationsintothe

auto-FAT10ylationofthebispecificE2conjugatingenzymeUBA6-specificE2 enzyme1.FEBSJ.281(7),1848–1859.

Amit,S.,Hatzubai,A.,Birman,Y.,Andersen,J.S.,Ben-Shushan,E.,Mann,M., Ben-Neriah,Y.,Alkalay,I.,2002.Axin-mediatedCKIphosphorylationof beta-cateninatSer45:amolecularswitchfortheWntpathway.GenesDev.16 (9),1066–1076.

Balkwill,F.,2009.Tumournecrosisfactorandcancer.Nat.Rev.Cancer9(5), 361–371.

Bates,E.E.,Ravel,O.,Dieu,M.C.,Ho,S.,Guret,C.,Bridon,J.M.,Ait-Yahia,S.,Briere,F., Caux,C.,Banchereau,J.,Lebecque,S.,1997.Identificationandanalysisofa novelmemberoftheubiquitinfamilyexpressedindendriticcellsandmature Bcells.Eur.J.Immunol.27(10),2471–2477.

Bett,J.S.,Kanuga,N.,Richet,E.,Schmidtke,G.,Groettrup,M.,Cheetham,M.E.,van derSpuy,J.,2012.TheinheritedblindnessproteinAIPL1regulatesthe ubiquitin-likeFAT10pathway.PLoSOne7(2),e30866.

Referenzen

ÄHNLICHE DOKUMENTE

Mammalian expression constructs for His 6 and Xpress-tagged human FAT10 (HisX- FAT10) and HA-tagged human NUB1L (HA-NUB1L) were gen- erated and transiently transfected into HEK293

Here, we show that the functionally barely characterized IFNc- and TNFa-inducible ubiquitin-like modifier FAT10 (also known as ubiquitin D, UBD), which binds to the autophagy

A yeast two hybrid screen identified NEDD8 ultimate buster-1long (NUB1L) as a non-covalent binding partner of FAT10 and this interaction was confirmed by coimmunoprecipitation and

Interestingly, ubiquitin and FAT10 turned out to be equally efficient at targeting long-lived proteins for degra- dation, thus indicating that FAT10 is the first ubiquitin-like

A version of NUB1L lacking all three UBA domains, on the other hand, looses the ability to bind FAT10 but is still able to interact with the proteasome and accelerates the deg-

As FAT10-expression in mTECs suggests a role in negative selection of T cells in the thymus, this was investigated in three distinct models: (1) Negative selection of

Since catalytic activity of HDAC6 is required for the transport of polyubiquitylated proteins to the aggresome (Kawaguchi et al., 2003) but is dispensable for the interaction with

Rather, similar to nonproteolytic roles of ubiquitin, SUMO modification has been shown to covalently modify a large number of proteins with important roles in many cellular