MitochondrialandMitochondrialDNAInheritance CheckpointsintheBuddingYeast, Saccharomyces cerevisiae DavidGarryCrider Submittedinpartialfulfillmentoftherequirementsforthe degreeofDoctorofPhilosophyintheGraduateSchoolof ArtsandSciences COLUMBIAUNIVERSITY 2012

©2011

DavidGarryCrider

Allrightsreserved

ABSTRACT

MitochondrialandMitochondrialDNAInheritance

CheckpointsintheBuddingYeast, Saccharomycescerevisiae

DavidGarryCrider

This dissertation analyzes the importance of mitochondria and mitochondrial DNA in

Saccharomyces cerevisiae during division . Movement and positional control of mitochondria and other are coordinated with cell cycle progression in the buddingyeast,Saccharomycescerevisiae.Recentstudieshaverevealedacheckpoint thatinhibitscytokinesiswhenthereareseveredefectsinmitochondrialinheritance.An establishedcheckpointsignalingpathway,themitoticexitnetwork(MEN),participatesin this process. Here, we describe mitochondrial motility during inheritance in budding yeast, emerging evidence for mitochondrial quality control during inheritance, and inheritancecheckpointsformitochondriaandotherorganelles.

TABLE OF CONTENTS

CHAPTER1...... 1

INTRODUCTION...... 1 MITOCHONDRIA...... 2 CELLCYCLE...... 2 CHECKPOINTS...... 5 MITOCHONDRIALINHERITANCE...... 10 QUALITYCONTROLDURINGMITOCHONDRIALINHERITANCE...... 11 MITOTICEXITNETWORKFUNCTIONINTHEMITOCHONDRIALINHERITANCE CHECKPOINT...... 16 CHAPTER2 ...... 20 MITOCHONDRIALINHERITANCEISREQUIREDFORMEN- REGULATEDCYTOKINESISINBUDDINGYEAST ...... 20 SUMMARY...... 21 RESULTSANDDISCUSSION...... 22 Mutationsthatinhibitmitochondrialinheritanceproducemultibuddedcellsinbuddingyeast....... 22 mdm10 ∆cellsexhibitdefectsincontractileringclosure....... 26 RolefortheMENinregulationofcellcycleprogressioninmdm10 ∆cells....... 32 ExperimentalProcedures...... 38 Yeaststrains,plasmids,andgrowthconditions:...... 42 Yeaststrainsusedforthisstudy...... 43 CHAPTER3...... 46

MtDNAINHERITANCECHECKPOINT...... 46 BACKGROUND...... 47 OTHERPROTEINSIMPLICATEDINmtDNAINHERITANCE...... 48 mtDNAMUTATIONS:rho 0ANDrho -CELLS...... 49 Rad53ANDTHEDNADAMAGECHECKPOINT...... 51 LINKSBETWEENRad53ANDmtDNA...... 54 RESULTS...... 56 LOSSOFmtDNAINDUCESAG1ARRESTINCELLCYCLEPROGRESSION...... 56

i

THEG1TOSPROGRESSIONDEFECTOBSERVEDINCELLSLACKINGmtDNAISNOTDUE TOLOSSOFMITOCHONDRIALRESPIRATORYACTIVITYORENERGYPRODUCTION...... 58 THEDEFECTINCELLCYCLEPROGRESSIONOBSERVEDINrho 0CELLSISDUETOLOSSOF DNAINMITOCHONDRIAANDNOTGENESENCODEDBYTHATDNA...... 61 ROLEFORAKNOWNCHECKPOINTPROTEIN(Rad53)INREGULATIONOFCELLCYCLE PROGRESSIONINCELLSLACKINGmtDNA...... 62 DISCUSSION...... 65 EXPERIMENTALPROCEDURES...... 68 Yeaststrains,plasmids,andgrowthconditions:...... 68 CHAPTER4...... 72

DISCUSSION...... 72 DISCUSSION...... 73 SUMMARYOFPROJECT1:...... 74 FUTUREEXPERIMENTS:...... 75 SUMMARYOFPROJECT2:...... 77 ROLEFORDNAPol γASASENSORFORTHEmtDNAINHERITANCECHECKPOINT:...79 HOWISTHESIGNALTHATmtDNALOSSTRANSMITTEDFROMMITOCHONDRIATO THENUCLEUS?...... 83 POSSIBLEROLEFORPROHIBITINSINTHEmtDNAINHERITANCECHECKPOINT...... 85 HOWMYSTUDIESMAYCONTRIBUTETOOURUNDERSTANDINGOF MITOCHONDRIALDISEASES...... 87 BIBLIOGRAPHY...... 90

ii

ACKNOWLEDGEMENTS

IwouldliketothankLizaPonforhermentorshipoverthepast3years.Thelab environmentshecreatedgiveseverygraduatestudenttheopportunityto succeed.Shesettheperfectbalancebetweenscientificcuriosityandreality;for whentofocusandfinishupthestory.Icannotthankherenoughforher dedicationandguidance.

ThePonlabwouldnotfunctionasefficiently,oraswellasitdoes,withoutIstvan

Boldogh.Heisthebackboneofthelab.Hehassuchawealthofknowledge,and suchaselflesspersonalitythathewilldropeverythingandhelpyouwheneverhe can.Iamsogratefulforallourconversationsandthetimewespenttogether.He trulymadethelabafunplace.

IwouldliketothankTomLipkinfortakingthetimetograbacupofcoffeeand encouragemetojointhePonlab.TheresaSwaynemightnotrealizethatshewas thefirstpersonthatItalkedtowhenIcametoColumbia(duringmyorientation) andI’mhonoredtobeabletothankheronmylastdayatColumbia.ToJose

“Ricky”RicardoMcFalineFigueroa(pickanameman!)whoislikeanolder,I meanyounger,brotherthatIneverhad,thankyou.Wediscussedverylittle scienceandforthatIcreditmycurrentsanity.ToLuisGarcia,whoIhavenever met,butwouldnothavehadsuchanexcitingprojectifitwasn’tforhisdiscovery andpreviouswork.

iii

Iwouldliketothankmyfirstscientificmentor,KennethBoheler,whoencouraged out-of-the-boxthinkingandmoldedmeintoascientist.Tothisdaymymost excitingexperimentwaswatchingmystemcellderivedcardiomyocytesbeaton thedish.WhowouldhavethoughtthatsingleexperimentwouldbethereasonI metmywife.I’dliketothankthoselittlecardiomyocytesforgrabbingmywife’s attentionandgivingmethechancetomeether,fallinlove,andgetmarried…my nextmostexcitingexperimentsareyettocome.

Myfamilyhasprovidedunconditionalsupportformethroughoutthelastdecade asIwouldfinishonedegreeandstartthenext.Theyneverquestionedmy dedicationandalwaysunderstoodandacceptedthesacrificethatittooktoreach mygoal.Icannevergetthoselostholidaysandtimespentawayfromthemback butIhopethesacrificeisjustifiedintheend.

Tomypap,thebestself-taughtscientistthatIknow.

Everyonementionedhadahugeroleintogettingmetothisdaybutnoone deservesmorecreditthanmywifeCheryl.SheisthereasonIcamebackto graduateschoolmorefocusedanddeterminedthanever.Icannotthankher enoughforherpatienceandunderstandingwhenIhadtoworkeveryweekend andstayinlablatebutalsoforthosedayswheresheputherfootdownand physicallydraggedmeoutoflab.SheknewwhenIwasburningmyselfoutand whentotellmetogetmyassoffthecouchandfinish.Ican’tdescribehowmuch youmeantomeandIcouldn’thavecompletedthisjourneywithoutyouandI cannotwaittostartthenextjourneywithyou.

iv

1

CHAPTER1

INTRODUCTION

2 MITOCHONDRIA

Mitochondria are essential organelles that perform fundamental cellular functions includingaerobicmetabolicfattyacidoxidation,aminoacidmetabolism,apoptosisand biosynthesis of many cellular metabolites. Mitochondria contain their own DNA

(mtDNA), which encodes respiratory chain components and tRNAs necessary for synthesis of mtDNA-encoded respiratory chain components, and cannot be made de novo. Saccharomyces cerevisiae has become an ideal model system for studying mitochondrialfunctionandinheritance,processesthatarerequiredforcellsurvival.The vital process of replicating and transferring ‘fit’ mitochondria to a new cell gives researchers a glimpse at the complex mechanisms involved in maintaining quality assurance for the next generation of cells. This thesis project will describe 2 major findingsonhowmitochondriaareregulatedthroughouttheentirecellcycleandhowthe presence and segregation of mitochondria and its genome in the daughter cell are criticalfornormalcelldivisiontooccur.Thefirstprojectinvestigateshowthepresence ofmitochondriainthedaughtercellisrequiredinordertocompletecytokinesis,andthe secondprojectexploreshowthepresenceofmitochondrialDNA(mtDNA)ismonitored andregulatedbyaconservedRad53checkpointsignalingpathway.

CELLCYCLE

RudolphVirhowhasbeencreditedfortheobservationin1855thatcellsariseonlyfrom pre-existingcells;whichlaterleadembryologiststodescribethecytologyofcelldivision in greater detail. However, the understanding of the underlying mechanisms of cell

3 division took place over a century later in the 1970s and 1980s where molecular biologists, cell biologists, biochemists, and geneticists joined forces to define and dissectthecellcycleinmolecularterms(Nurseetal.,1998).Theirworkrevealedthe basicstructureandcomponentsofcelldivision,leadingtotheunderstandingthatthis eventishighlyregulated.

The cell cycle is a series of events and processes that lead to the replicating and segregationofessentialorganellestoanewcellduringcellcycledivision.Thecellcycle isoftencomposedoffourphases,thetimebeforeDNAreplicationisknownas(G1),the

DNAsyntheticphase(S),theprocessafterDNAreplication(G2),andthemitoticphase

(M),whichiswherecellssegregateduplicatedchromosomes(Figure1.1).Theconcept of rate-limiting steps during known phases of the cell cycle lead to the discovery of maturation promotion factor (MPF) in fission yeast mutants, (later found in starfish oocytes,blastomeresoffrogembryosandinhumancells),thatprematurelyadvanced toandeventuallyledtotheobservationofcyclindependentkinases(CDKs)as regulators of the cell cycle (Nurse et al., 1998). Eukaryotic cells commit to each new cycle in mid-G1. In yeast, this occurs before bud emergence. This commitment or transitionisreferredtoasthe‘restrictionpoint’or‘START’.

Progressing beyond ‘START’ is the major target for growth factors and nutritional signalingandoccursbyactivationofG1phasecyclin-dependentkinases(CDKs).Italso marksthetimeinwhichthereplicationofthenucleargenome,anerrorproneprocess, begins.Errorswithinthegenomemustbeidentifiedandcorrectedbeforethegenomeis

4 inheritedtothenextgenerationofcells.Therefore,itisnotsurprisingthatcellshavea quality control mechanism that induces a transient arrest in G1 if DNA damage is detected in order to repair mutagenic DNA damage before replication occurs in S- phase.

DNA monitoring is one example of a regulatory surveillance and quality control apparatus that is in place to limit and reduce erroneous DNA to be inherited by the daughter cell. Eukaryotic cells also cannot divide until the genomes (DNA) are replicatedandtransferredtothenewdividingcells.Thiscontrolduringcelldivisionis notlimitedtojustgenomicmaterial;theisalargemacromolecularstructure that moves and assemblies and these events are also under surveillance during all stagesofthecelldivision.Indeed,celldivisionisunderconstantsurveillanceandthe initiation of late events is dependent on the completion of early events. Dependent relationshipsseeninsomaticcellsareakeyelementinunderstandingthehighfidelity of organelle reproduction and distribution during cell division (Hartwell and Weinert,

1989). Saccharomyces cerevisiae , budding yeast, has been instrumental in the identification and characterization of regulatory components during successful cell divisionsaswellasidentifyingkeyproteinsinvolvedinthisregulatoryprocess.

5

Figure1.1General OutlineoftheCell CycleinBudding Yeast. Thecell commitstoaroundof celldivisiononceit passesthe‘transition orStart’pointinmid G1.Thereplicationof thenucleargenome takesplaceduringS Phaseandnuclear segregationshortly followsthereplication duringG2.Organelle duplicationand migrationiscompleted inmitosisand mpf.biol.vt.edu. Budding yeast homepage cytokinesismarksthe endofonedivision andthebeginningof thenext.

CHECKPOINTS

Eukaryoticcellsdevelopedacomplexnetworkofsignalpathwaystoensurehighfidelity replication and transfer of macromolecules from mother cell to daughter cell. This processishighlycontrolled:bothintrinsicandexternalsignalscanactivatearegulatory signal known as a checkpoint. This regulation is a control mechanism called a checkpoint.Checkpointsareinplaceto‘check’thatcriticaleventsofthecellcycle,such as genomic and macromolecular assembly and organization, are properly executed.

Theconceptof‘checkpoints’arosefromthediscoveryofgenesinbuddingyeastthat

6 regulate the process of cell cycle events and have the ability to delay or block the continuationofcelldivisioniferrorsarefound(HartwellandWeinert,1989).

Checkpointscontrolthetimingandcoordinationofeventsduringcelldivisionandwere originallyidentifiedinbuddingyeast(HartwellandWeinert,1989).Hartwelldiscovered and generated a collection of cdc (cell-division cycle) mutants in which Hartwell and others were able to isolate key cell cycle genes (Hartwell et al., 1970). This further showedtheabilityandpowerofgeneticstostudyanddefinecellcycleregulators.Yeast providedanidealorganismtostudythe cdc mutantscollectiongeneratedbyHartwellby geneticallyandvisuallyobservingspecificphasesofthecellcycleinordertomapout themechanismsinvolved.Aremarkablefindingwasthatmost cdc genesareconserved and have homologues in all eukaryotes and have similar key regulating functions in speciesrangingfromyeasttohumans(Hartwelletal.,1970) (Nurseetal.,1998).An important early finding by Hartwell was the identification of cdc28 as the gene that controlsSTART.ThisCDKgenewasamajordiscoverythatformedthebasisofour understandingofthecellcycle(Nurseetal.,1998).

Latecellcycleeventsarethoughttobedependentonearliereventsbyeithersubstrate- productpathwayoracontrolmechanismbutcellcycleeventsthatarenot‘hardwired’ together in the same manner as metabolic pathways and, therefore, they can be mutatedandfurtherstudiedanddissected(HartwellandWeinert,1989) (Nurseetal.,

1998).Theseeventsandphasescanbestudiedbymutantsthatspecificallyinhibitone event of the cell cycle. Temperature sensitive mutants in S. cerevisiae exist for bud

7 formation, spindle pole body enlargement, spindle elongation, initiation of DNA

(replication,elongation,andligation),chromatinassembly,chromosomeassemblyand segregation, nuclear division, and cytokinesis (Hartwell and Weinert, 1989). The existenceofcontrolmechanismsaresupportedbyconditionsthatpermitlateeventsto occur even when an early normal prerequisite event is prevented, a term coined by

Hartwellas‘reliefofdependence’(HartwellandWeinert,1989).Reliefofdependency experimentsidentifiedanumberofcheckpointscontrolmechanisms.

Hartwelletal.identifiedaregulatorycheckpointcontrolmechanismformakingmitosis dependent on the completion of DNA replication (Hartwell and Weinert, 1989).

Coordinated eventsallow the stoppage of cell division during multiple stages if errors aredetected.Thedelaysaremediatedbygeneticallyencodedcheckpointcontrolsthat arenotaconsequenceofthedamageperseandarenotessentialforcellcycleevents

(e.g.DNAreplicationormitosis).Instead,checkpointsonlyinitiateadelayafterdamage is detected (Weinert, 1998). Perturbation of DNA replication by inhibitors or by mutations,resultsininactivationofreplicationenzymes,whichinturnpreventspassage through mitosis in yeast and many other eukaryotic organisms. cdc9 temperature sensitive mutants were blocked at mitosis and failed to bud when grown at the restrictive temperature but cells could proceed past mitosis when they contained an addition rad9 mutant.Suggesting RAD9 geneanditscomponentscontrolthissystem

(HartwellandWeinert,1989).

Further, a role for RAD9 in cell cycle progression in response to defects in DNA

8 synthesiswasdeterminedusingacollectionoftemperaturesensitivemutantsthatwere initially identified by failure to arrest the cell after DNA damage was induced by x- irradiation (Hartwell andWeinert, 1989). Mutations in the RAD9 gene allow cells with

DNAdamagetoproceedthroughcelldivision,whereasirradiatedwild-typecellsarrest in G2 until the DNA damage is repaired. This was illustrated by relieving the dependency of mitosis on the completion of DNA synthesis through the use of temperaturesensitivemutantsinvolvedinDNAreplicationandbyknockingout RAD9 whichenabledthecellstocontinuethroughmitosisintothenextcellcycle(Hartwelland

Weinert,1989).Cellcycleprogressionwasaccessedbyvisualizationofbudsizeand analysis of DNA content either through DAPI staining orflow cytometry. Temperature sensitivemutantswereusedtoconfirmthecontrolofcellcycleprogressionby RAD9 monitoring components of DNA replication. This illustrates that a control pathway,

RAD9, mustbeactiveinordertoarrestDNAreplicationdefectivecellsbeforemitosis andisthoughttobethemainprinciplethatappliestomanycheckpoints.

Checkpointsandotherqualitycontrolmechanismsincreasethefidelityofcelldivision by triggering pathways to repair errors or to cause cell death if the error cannot be repaired. DNA damage cascades are linked to at least 3 checkpoints: G1/S (G1) checkpoint,intra-Sphasecheckpoint,andG2/Mcheckpoint.However,qualitycontrolis notlimitedtojusttheintegrityofDNA.Therearecheckpointsthatmonitorthepresence ofspecificstructuressuchasspindleformationandlocalization.Thischeckpoint,known asthespindlecheckpoint,illustratestheimportanceofpolaritythroughouttheentirecell cycle. Upon activation, the spindle checkpoint arrests cell cycle at M phase until all

9 chromosomesarealignedonthespindle.Anothercytoskeletalcheckpointisreferredto asthemorphogenesischeckpointandhasalsobeenidentifiedinyeast.Thischeckpoint detects abnormality in the and arrests the cell cycle at G2/M transition.

Thesecheckpointsensurethatgenomicintegrity,replication,andsegregationworkin harmonywithmechanismsunderlyingmovementandlocalizationofsegregatingcellular constituents. Saccharomyces cerevisiae has been invaluable in dissecting the mechanisms involved in identifying the series of eventsfrommoving macromolecules fromonecelltothenextduringcelldivision.

The elimination of checkpoints may have both a subtle or catastrophic consequence dependingonprevailingconditions.Thehumanhomologuesofseveral S.cerevisiae checkpoint genes map to chromosomal regions implicated in the etiology of a wide variety cancers. Specifically, Rad9 expression has been associated with prostate, breast,lung,skin,thyroidandgastriccancersandhighexpressionhasbeenassociated withhumanprostatecancergrowth(BroustasandLieberman,2011).Also,thedeletion of Rad9 in mouse models shows a higher incidence of skin cancer, therefore, it is thought that Rad9 can act as an oncogene or tumor suppressor (Broustas and

Lieberman,2011).Controlofcellcyclephaseprogressionandthelinktocancerisnot limitedto Rad9 only;manycheckpointproteinssuchas ATM,ATR,Chk1, and Chk2 are major signaling molecules that are involved with both endogenous and exogenous sources of DNA damage. Many cell cycle protein mutations are thought to have fundamentalrolesinthepathogenesisofhumancancers(DaiandGrant,2011;Hartwell andWeinert,1989) (HartwellandKastan,1994).

10

Theunderstandingofcheckpointproteinscouldshedlightonthemechanisminvolved incancerdevelopmentandprovidetherapeutictargets.Further,Cellcyclecheckpoints areimportantintheprotectionandabilitytorescuethecellsfromDNAdamageinduced bycurrentchemotherapeuticagentsandradiationtherapy(DaiandGrant,2011).Yeast remains an ideal organism to study checkpoint proteins because of large percent of homologybetweenhumanandyeastcheckpointmechanismsandproteins.

MITOCHONDRIALINHERITANCE

In early characterizations of mitochondrial morphology and distribution mutants, Sogo andYaffe(SogoandYaffe,1994)notedamultibuddedphenotypeinyeastbearinga mutation in MDM10 ,nowknowntoencodeacomponentofthemitochore. My thesis workrevealedthatthemultibuddedphenotypeobservedin mdm10 ∆occursasaresult of a mitochondrial inheritance checkpoint: a mechanism that inhibits cell cycle progressionatcytokinesiswhentherearedefectsinmitochondrialinheritance(Garcia-

Rodriguez et al., 2009). I also found that a known cell cycle checkpoint signaling pathway,theMitoticExitNetwork,regulatesthemitochondrial inheritance checkpoint.

Below,Idescribetheeventsthatoccurduringmitochondrialinheritanceinyeast,and the mechanisms underlying mitochondrial movement, immobilization and segregation duringyeastcelldivision.

11

QUALITYCONTROLDURINGMITOCHONDRIALINHERITANCE

Qualitycontrolandtheestablishmentofsignalingcascadestoensurepropertemporal eventsisnotlimitedtojustthegenome.Manydiverse mechanisms have evolved in eukaryotic organisms for the inheritanceoftheir organelles upon cell division. These mechanismsdependonthecharacteristicsofagivenorganelle,suchasitsstructure, abundanceandarrangementinthecell,andarecoupledtothenatureoftheprocessby whichacelldivides.

Celldivisionin S.cerevisiae takesplacebyanasymmetricprocess. S.cerevisiae grow anddivideasymmetricallybybuddingand,therefore,theinheritanceoftheirorganelles frommothertodaughtercellsreliesonanactivesegregationprocess.Thisprocessis highlyregulatedandcoordinatedwiththeprogressionofthecellcycle.Althougheach organelle is transferred to the bud by a different mechanism, there are important common strategies used by yeasts to inherit organelles (for review see (Fagarasanu andRachubinski,2007).

As illustrated in the case of mitochondria (Fig. 1.2), a fraction of the organelles are engagedinlinearpolewardmovementstowardsthebud(anterogrademovement)and awayfromthebud(retrogrademovement).ThismovementisinitiatedduringSphase ofthecellcycle,whenthebudemerges,andcontinuesuntiltheendofthecelldivision cycle(BoldoghandPon,2007).Inaddition,somemitochondriaareimmobilizedatthe distaltipofthemothercell,ensuringthatnotallorganellesaretransferredintothebud.

12

Othermitochondria,whichhavebeentransportedto thebudtip,areanchoredthere.

Anchorageofmitochondriainthebudtipensuresthattheseorganellesareretainedin thebud.Polewardmitochondrialmovement,togetherwithanchorageoftheorganelleat thepoles,resultsintheequalpartitioningofmitochondriabetweenmotheranddaughter cellsduringyeastcelldivision.

Figure 1.2. The mitochondrial inheritance cycle in budding yeast. Mitochondria align along the mother-bud axis and orient toward the site of bud emergenceatG1.DuringS,G2,andmitosis,mitochondriamovelinearlytowardthetip ofthebudormothercell.Mitochondriaareimmobilizedatthebudtipormothercelltip until the end of the cycle, when they are released and redistributed throughout the .

13

Segregation of mitochondria is a complex process that relies on a large number of proteins with diverse functions. The movement and segregation of mitochondria and other organelles in S. cerevisiae depends on the actin cytoskeleton. Therefore, a significant proportion of these proteins are involved in structuring and remodeling the actincytoskeleton.

In yeast, there are two F-actin-containing structures that persist throughout the cell division cycle: actin patches and actin cables (reviewed in ref. (Moseley and Goode,

2006).Actinpatcheswerenamedfortheirappearance in phalloidin-stained cells; but theyareactually,investedwithanF-actin coat, that form in the bud and budtip(Fehrenbacheretal.,2004).ActincablesarebundlesofF-actinthatcontainthe actin-bundling proteins fimbrin (Sac6p) and Abp140p, and two tropomyosin isoforms

(Tpm1p and Tpm2p). Formin proteins, Bni1p and Bnr1p, mediate nucleation and elongationofF-actinfilamentsthatarethenbundledintoactincables.Bni1pandBnr1p localizetothebudtipandbudneck,whicharethesitesofactincableassembly.

Actincablesextendfromtheirassemblysitesalongthemother-budaxisofthecelland arealsodynamicstructuresthatundergoretrogrademovementfromthebudtowardthe mothercell(Fig.1.3A)(YangandPon,2002).Retrogradeactincableflowisdrivenin partbyactincableassemblyandelongation,whichoccurscontinuouslyandprovidesa pushingforceforretrogradeflow.AtypeIImyosinatthebudneck,Myo1p,provides pullingforceduringretrogradeactincableflow(Huckabaetal.,2004)(Huckabaetal.,

2006).

14

Figure 1.3. Model for actin-driven

bidirectionalmovementofmitochondriain

budding yeast. Actin cables are bundlesof

F-actinthatalignalongthemother-budaxis.

Anterogrademitochondrialmovement,toward

thebudtip,andretrogrademovementofthe

organelle,towardthemothercelltip,areboth

dependent on actin cables. The mitochore

mediates reversible binding of mitochondria

and mitochondrial DNA to actin

cables for anterograde and retrograde

movement. (A) Anterograde mitochondrial

movement is driven by the Arp2/3 complex,

which is recruited to the mitochondrial

surfacebyJsn1p/Puf1pandstimulatesforce generationforbud-directedmovementalongactincablesthroughactinpolymerization.

Puf3p promotes anterograde mitochondrial movement by linking the mitochore and mitochondria associated Arp2/3 complex. (B) Retrograde mitochondrial movement is driven by the retrograde flow of actin cables, whichisdrivenby thepushofformin- stimulated actin polymerization and assembly into actincablesandthepulloftypeII myosins.Thus,mitochore-mediatedbindingofmitochondriatoactincablesundergoing retrogradeflowlinkstheorganelletoits‘conveyorbelt’forretrogrademothertipdirected movement.

15

The essential function of actin cables in budding yeast is to drive intracellular movements that are required for bud growth and organelle inheritance. Mitochondria undergo reversible binding to F-actin in vitro, require actin cables for movement and inheritance,andundergoanterogradeandretrogrademovementalongactincablesin vivo (Lazzarino et al., 1994) (Simon et al., 1997) (Fehrenbacher et al., 2004).

Associationofmitochondriawithactincablesforanterogradeandretrogrademovement requires a mitochondrial outer membrane protein complex, the mitochore, which consists of the proteins Mdm10p, Mdm12p, and Mmm1p (Boldogh et al., 1998)

(Boldogh et al., 2003). For retrograde movement, mitochondria undergo mitochore dependentbindingtoactincables,andusetheforcesofretrogradeactincableflowto drivetheirmovementtowardthedistaltipofthemothercell(Fig.1.3B)(Fehrenbacher etal.,2004).

Foranterogrademovementofmanyorganelles,propulsionisprovidedbymyosinmotor proteinsoftheclassVfamilyofmyosins,whichmoveandtransportcargoestowardthe barbedendsofactinfilamentswithinactincables.In S.cerevisiae ,therearetwoclass

V myosins: Myo2p, which transports secretory vesicles, , , and lateGolgivesicles;andMyo4p,whichtransportsthecorticalER(cER,seebelow)and mRNA(Fagarasanuetal.,2007).

Myo2p can bind to mitochondria in vitro and is required for normal mitochondrial distribution(Altmannetal.,2008)(Itohetal.,2004)(Itohetal.,2002).However,live-cell imaging and biochemical evidence implicate an alternative propulsion mechanism in

16 which the mitochore mediates association of mitochondria with actin cables, and the

Arp2/3 complex generates force through actin polymerization for movement of mitochondriaalongthecables(Fig.1.3A)(Boldoghetal.,2001)(McKaneetal.,2005)

(SenningandMarcus,2010).TheArp2/3complexassemblesF-actinbybindingtothe sideofapreexistingfilamentandstimulatingnucleationofanewfilamentata70°angle relativetothepre-existingfilament(recently reviewedinref.(CampelloneandWelch,

2010). Jsn1p, a Pumilio family protein that localizes to the mitochondrial outer membrane,isanArp2/3complexreceptoronmitochondria(Fehrenbacheretal.,2005).

Puf3p,anothermitochondria-associatedPumiliofamilyprotein,mediatesassociationof the Arp2/3 complex with the mitochore, which coordinates anterograde forces generatedonmitochondriawithassociationoftheorganellewithactincables(Garcia-

Rodriguezetal.,2007).

MITOTICEXITNETWORKFUNCTIONINTHEMITOCHONDRIAL INHERITANCECHECKPOINT

When I began my thesis research, it was clear that mitochondria undergo cell cycle linked changes in position and movement, which ensure equal segregation of the organelle between mother and daughter cells. Indeed, segregation of mitochondria during cell division in budding yeast exhibits features that resemble chromosome segregation: both undergo poleward movement followed by anchorage at the poles.

Moreover, there are many known checkpoints that inhibit cell cycle progression and activate cellular repair pathways when there are defects in chromosome duplication

17 and/or segregation. However, there are no known checkpoints for inheritance of organellesotherthanthenucleus.

My thesis research revealed a mitochondrial inheritance checkpoint that inhibits cytokinesiswhentherearedefectsinmitochondrialinheritanceinbuddingyeast,anda novel role for the MEN in this process. My thesis research also revealed a mtDNA inheritance checkpoint that inhibits G1-to-S progression in response to defects in mtDNAinheritance,andarolefortheDNAdamagecheckpointinthisprocess.Below,I describe the MEN. In the introduction to Chapter 3, I described the DNA damage checkpoint,andfunctionallinksbetweenthispathwayandmtDNA.

The mitotic exit network (MEN) is a GTPase-driven signal transduction cascade that was originally identified for its role in coordinating chromosome segregation and exit from mitosis, and ensuring proper segregation of genetic information (Krapp et al.,

2004;SeshanandAmon,2004).MENregulatorsareconservedinyeasts,C.elegans, and mammalian cells (Fig. 1.4). The central players in the MEN are the protein phosphatase Cdc14p, the small G protein Tem1p, and Lte1p and Bub2p/Bfa1p, the activator and GAP/inhibitor, respectively, for Tem1p. Activation of Tem1p, which ultimatelyleadstoactivationofCdc14pandlocalizationofactiveCdc14ptoitssitesof action,isrequiredformitoticexitandcompletionofcytokinesis.

18

SeveralstudiesindicatethattheMENalsohasaroleinregulatingcytokinesisinyeast.

First, several MEN components localize to the site of contractile ring assembly (Luca andWiney,1998)(Frenzetal.,2000)(Songetal.,2000)(Xuetal.,2000)(Yoshidaand

Toh-e,2001)(Bembeneketal.,2005).Second,conditionsthatbypassMENfunctionin mitotic exit (including mutations that weaken interactions of Cdc14p with its inhibitor

Cfi1p/Net1p,overexpressionoftheCDKinhibitorSic1p,ormutationsthatinhibitexport ofactiveCdc14pfromthenucleustothe)produce severe cytokinesis defects

(Songetal.,2000)(Bembeneketal.,2005)(Jimenezetal.,1998)(Shouetal.,1999)

(Lippincott et al., 2001) (Hwa Lim et al., 2003). Third, recent studies indicate that the

MENmaycontrolcytokinesisbytargetingtheproteinsimplicatedinseptaformationto thebudneck(Blondeletal.,2005)(Meitingeretal.,2010)(Nishihamaetal.,2009).

Figure1.4Cdc14pisregulatedby FEAR andMEN signaling pathways. During early anaphase the FEAR (for Cdc-fourteen early anaphase release) signalingphosphatase,Cdc14p,issequesteredinthe byaninhibitorcomplexCfi1p/Net1p.When correct spindle orientation occurs in early anaphase, some Cdc14p is activated and released from the nucleolus by the FEAR pathway (Dimmer et al., 2005). Further activation of Cdc14 occurs in late anaphase by the MEN. Tem1p, a small GTPase protein in the MEN pathway, is negatively regulated by a GTPase-activating protein (GAP) complex Bub2p-Bfa1p,whichisnegativelyregulatedbyLte1p and Cdc5p. Activated, GTP-bound Tem1p then initiates a signaling cascade by activation of the protein kinase Cdc15p. Cdc15p then activates the Dbf2p/Mob1p protein kinase complex, which further activates Cdc14p allowing for progression through mitoticexitandcytokinesis.

19

MythesisresearchsupportsarolefortheMENininhibitingcytokinesisinresponseto defectsinmitochondrialinheritance.Thesestudies provide additional evidence for a role of the MEN in regulating cytokinesis, independent of its function in regulation of mitotic exit. They also reveal a broader surveillance function for the MEN than previouslyobserved.

20

CHAPTER2

MITOCHONDRIALINHERITANCEISREQUIRED FORMEN-REGULATEDCYTOKINESISIN BUDDINGYEAST

21

SUMMARY

Mitochondrial inheritance, the transfer of mitochondria from mother to daughter cell duringcelldivision,isessentialfordaughtercellviability.Themitochore,amitochondrial protein complex containing Mdm10p, Mdm12p and Mmm1p, is required for mitochondrialmotility,leadingtoinheritanceinbuddingyeast.Weobserveadefectin cytokinesis in mitochore mutants and another mutant ( mmr1 ∆ gem1 ∆) with impaired mitochondrial inheritance. This defect is not observed in yeast that have no mitochondrial DNA or defects in mitochondrial protein import or assembly of β-barrel proteins in the mitochondrial outer membrane. Deletion of MDM10 inhibits contractile ring closure, but does not inhibit contractile ring assembly, localization of a chromosomal passenger protein to the spindle during early anaphase, spindle alignment,nucleolarsegregationornuclearmigrationduringanaphase.Releaseofthe mitoticexitnetwork(MEN)component,Cdc14p,fromthenucleolusduringanaphaseis delayed in mdm10 ∆ cells. Finally, hyperactivation of the MEN by deletion of BUB2 restoresdefectsincytokinesisin mdm10 ∆ and mmr1 ∆ gem1 ∆cells,andreducesthe fidelityofmitochondrialsegregationbetweenmotheranddaughtercellsinwild-typeand mdm10 ∆cells.OurstudiesidentifyanovelMEN-linkedregulatorysystemthatinhibits cytokinesisinresponsetodefectsinmitochondrialinheritanceinbuddingyeast.

22 RESULTSANDDISCUSSION

Mutationsthatinhibitmitochondrialinheritanceproducemultibudded cellsinbuddingyeast.

Equalsegregationofmitochondriabetweenmotheranddaughtercellsduringyeastcell divisionoccursasaresultofbidirectionalmovementofmitochondriatothebudtipand mother cell tip and anchorage of the organelle at those sites (Fehrenbacher et al.,

2004). The mitochore, a mitochondrial membrane protein complex containing the proteinsMmm1p,Mdm10pandMdm12p,isrequiredforbindingofmitochondriatoactin filaments in vitro , actin cable-dependent bidirectional mitochondrial movement, and mitochondrialinheritance(Fehrenbacheretal.,2004)(Boldoghetal.,1998)(Boldoghet al., 2003). In early characterizations of mitochondrial morphology and distribution mutants,SogoandYaffe(SogoandYaffe,1994)notedthepresenceofamultibudded phenotypein mdm10 ∆cells.Wefindthatmultibuddedclustersconsistingof3-5buds are present during mid-log phase and accumulate with growth time in mdm10 ∆ cells.

This multibudded phenotype is observed in mdm10 ∆ cells in three different genetic backgrounds:S288C,W303andA264A(datanotshown).

Inwild-typeyeast,mitochondriaconstituteadynamicandtubularreticulum(Fig.2.1A-B)

(Fehrenbacher et al., 2004). In mdm10 ∆ cells, mitochondria are large spherical structures that fail to move from mother cells to buds and undergo rapid loss of mitochondrial DNA (mtDNA) (Boldogh et al., 1998; Boldogh et al., 2003). The large spherical mitochondria typical of mdm10 ∆ cells are usually present in only one cell

23 withinamultibuddedclump(Fig.2.1E-F).VisualizationofDNAconfirmedthat mdm10 ∆ cellshavenomtDNAandrevealedthateachcellbodyin mdm10 ∆clumpscontainsa nucleus(Fig.2.1G-H).Theviabilityofwild-typeand mdm10 ∆cellsduringmid-logphase growth, assessed using FUN-1 staining, is 93.5% and 76.5%, respectively. Thus, a mutation in MDM10 that results in severe defects in mitochondrial morphology and inheritance also produces defects in mother-daughter cell separation but does not inhibitnuclearinheritanceorcompromisecellviabilityinSCmedium.

Deletionof MDM10 , MDM12 or MMM1 alsoresultsindefectsinmaintenanceofmtDNA, mitochondrialmorphologyandassemblyof β-barrelproteinsinthemitochondrialouter membrane(OM)(Boldoghetal.,1998;Hobbsetal.,2001;Meisingeretal.,2004;Sogo andYaffe,1994).Therefore,wetestedwhetherthemultibuddedphenotypeof mdm10 ∆ cells is due to defects in these mitochondrial inheritance-independent processes by analysis of yeast bearing deletions in mtDNA, MAS37 or TOM7 . rho 0 cells have no mtDNAandseveredefectsinmitochondrialrespiration(Goldringetal.,1970).Mas37p isasubunitoftheSAM/TOBcomplex,whichmediates assembly of β-barrel proteins intothemitochondrialOM(Wiedemannetal.,2003).Tom7pisasubunitoftheprotein- translocatingporeinthemitochondrialOM(Honlingeretal.,1996).Deletionof TOM7 produces defects in mitochondrial morphology that are similar to those observed in mdm10 ∆cellsaswellasdefectsinmitochondrialproteinimport(Meisingeretal.,2004).

Tom7p also promotes the segregation of Mdm10p from the SAM/TOB complex

(Meisingeretal.,2006).

24

rho 0, mas37 ∆ and tom7 ∆ cells exhibit significantly lower defects in mitochondrial inheritanceandlowerlevelsofmultibuddedcellscomparedtomitochoremutants(Fig.

2.1 I-J). Thus, the multibudded phenotype observed in mdm10 ∆ cells is not a consequence of loss of mtDNA, or of defects in mitochondrial respiratory activity, proteinimport,orOM β-barrelproteinassembly.Moreover,weobservedalinkbetween the extent of multibudded cells in late-log phase cultures and the severity of the mitochondrial inheritance defect in yeast carrying mutations in mitochore subunits: mdm10 ∆ = mmm1- 1 > > mdm12 ∆ (Fig. 2.1 I-J). Mdm12p coordinates mitochondrial inheritance and biogenesis through its direct interactions with the PUF family protein

Puf3p (Garcia-Rodriguez et al., 2007). Thus, mdm12 ∆ cells may have less severe multibudded and inheritance phenotypes compared to mdm10 ∆ or mmm1-1 mutants becauseMdm12phasregulatoryeffectsonmitochondrialmotility,while Mdm10pand

Mmm1p have predominant roles in mediating mitochondrial motility. Overall, the multibudded phenotype observed in all mutants analyzed correlates with defects in mitochondrialinheritance.

25

Figure2.1.Cellseparationdefectsinmitochondrialinheritancemutants. Wild-type(A-D) (BY4741)ormdm10 ∆(E-H)(398)cellsweregrowninSCmediumat30°Ctomid-logphase. Cells were either stained for mitochondria using MitoTracker Red (MtRed), or fixed with formaldehydeandstainedusingtheDNA-bindingdyeDAPI.ImagesofMtRedorDAPIstained cells are 2-D projections of the reconstructed 3-D volume that are superimposed on the correspondingphaseimage.(A-BandE-F)PhaseimagesandMtRedstainingofwild-typeand mdm10 ∆ cells, respectively. The arrow points to the original mother cell in a multibudded mdm10 ∆clusterthatcontainsalargespherical(F).Bar:1µm.(C-DandG-H) PhaseimagesandDAPIstainingofwild-typeandmdm10 ∆cells,respectively.n:nuclearDNA. m: mtDNA. (I-J) Quantification of mitochondria-free buds in cell bearing small buds (I) and multibudded cells (J) in wild-type (ISY001), rho0 (ISY001-rho0), mas37 ∆ (ISY005), tom7 ∆ (ISY006), mdm12 ∆ (ISY003), mmm1-1 (ISY065) and mdm10 ∆ (ISY002) cells (n>800). Cells weregrowninSCmediumat30°Cfor12-16hrstolate-logphase(OD600=1.2–1.4).Error barsarestandarddeviations.

26 mdm10 ∆cellsexhibitdefectsincontractileringclosure. mdm10 ∆cellsthatenterthecellcyclehaveasignificantdelayintheabilitytoenterG2 phasecomparetowild-typecells(Fig.2.2).Spindleassemblyanddisassemblyaswell astheappearanceanddisappearanceofmitoticcyclinaredelayedtoasimilarextentin mdm10 ∆ compared to wild-type cells (Fig 2.3). Formation of the second bud (d2) in multibudded mdm10 ∆cellsoccurs150minafterreleasefrompheromone-induced G 1 arrest, 25 min after the first bud (d1) undergoes Clb2p degradation and spindle disassembly(Fig2.3).

27

Fig 2.2. mdm10 ∆ cells exhibit a delay in progression from G 1 to G 2. Wild-type (ISY001), mdm10 ∆(ISY002)orrho 0(ISY001-rho 0)yeastweregrowninSCmediumat 0 30 Ctomid-logphase(OD 600 =0.5–0.8).Cellswereincubatedwitha-factor(10µM) for2.5hrstoinducearrestinG 1 phase.Thereafter,theywerewashedwithpheromone- free SC medium and incubated at 30°C. At various times min after release from pheromone-induced G 1 arrest, cells were fixed, stained with propidium iodide and analyzedforDNAcontentbyflowcytometry.ThepercentageoftotaldividingcellsinG 2 phase,asafunctionoftimeafterreleasefrompheromone-inducedG 1 arrest,isshown. The basis for the reduced rate of progression of mdm10 ∆ cells from G 1 to G 2 is not clear.However,rho 0 cells,whichhavenomtDNAandseveredefectsinmitochondrial respiratoryactivity,canprogressthroughtheG2 phaseatasimilarrateasthatofwild- typecells.Thus,thereducedrateofcellcycleprogressionobservedin mdm10 ∆cells doesnotappeartobelinkedtomitochondrialrespirationordependentuponmtDNA.

28

Fig2.3mdm10 ∆cellsexhibitadelayanaphaseentryandmitoticexit .A)Awild- typestrain(BY4741)andanmdm10 ∆mutantstrain(ISY002)weresynchronizedasfor Fig.2.2andgrownat30°C.Aliquotswereremovedfromculturesatthetimeindicated. LevelsofClb2pinsynchronizedcellculturesweredeterminedbyWesternblotanalysis usingapolyclonalanti-Clb2pantibody.Hxk1pwasusedasaloadingcontrol.B)Wild- type(LGY020;blackline)andmdm10 ∆(LGY021;greyline)cellsexpressingplasmid- borne mCherry-tagged were synchronized and aliquots were removed from culturesatthetimesindicated,fixedandvisualizedbyfluorescencemicroscopy.The number of cells with spindles > 4 µm in length was determinedasafunctionoftime after release from pheromone-induced G1 arrest. Spindle assembly and disassembly are also delayed in mdm10 ∆ compared to wild type cells. We detect the maximum numberofanaphasespindles,spindlesthatare4-7 µm in length, within 60 min after releaseofwild-typecellsfrompheromone-inducedG1arrest,andspindledisassembly 100minafterreleasefromG1arrest.Thekineticsofanaphasespindleassemblyand disassembly in these synchronized wild-type cells are similar to those reported previously(Goldringetal.,1970).Incontrast,deletionofMDM10resultsina25min delayinspindleassemblyanda35mindelayinspindledisassembly.Accountingfor thedelayinanaphaseonset,mdm10 ∆cellsexhibita10mindelayinmitoticexit.The delayinearlyanaphaseandmitoticexitobservedinmdm10 ∆issimilartothedelayin G1toG2phaseprogression.C)Quantitationofthetimecourseforformationofsecond budsinmultibuddedmdm10 ∆cells.

29

Interestingly, rho 0 cells undergo a G1 delay in cell cycle progression similar to that observedin mdm10 ∆;therefore,thedecreaseincellcycleprogressionin mdm10 ∆may beduetolossofmtDNA. However,themultibuddedphenotypein mdm10 ∆cellsisnot duetolossofmtDNA(Fig.2.1J),ortodefectsinseptation(degradationofthecellwall betweenmotheranddaughtercells)(datanotshown).Spindlealignmentwasconfirmed tobenormalbylookingattubulinstainingandnucleolarsegregationwasvisualizedby the visualization of a nucleolus protein, nop1 (data not shown). Rather, it is due to defectsincontractileringclosure.Actomyosinring contraction was visualized in wild- type and mdm10 ∆ cells using afully-functionalfusion protein consistingofthetypeII myosin(Myo1p)fusedtoGFP(LippincottandLi,1998),mitochondria-targetedDsRed, and4-Dimaging(timelapseimagingcombinedwith3-Dreconstruction).

Deletionof MDM10 hasnoeffectoncontractileringassembly:Myo1p-GFPlocalizesto a ring at the mother-bud junction in both wild-type and mdm10 ∆ cells (Fig. 2.4 A-D).

Moreover, mdm10 ∆cellshavethecapacitytoundergocontractileringclosure(Fig.2.4

B),andtodosowithkinetics(14.2±3.5min,n=48)similartothatofwild-typecells

(10.4±2.1min,n=43).Thereissomelossofsynchronyin mdm10 ∆cellsatthetimeof contractile ring closure. Nonetheless, mdm10 ∆ cells that undergo contractile ring closuredoso20-40minlaterinthecellcyclecomparedtowild-typecells(n=48).

However, mdm10 ∆cellsexhibitdefectsincontractileringclosure,whichcorrelateswith defects in mitochondrial inheritance (Fig. 2. 4 C). To quantitate the frequency of contractileringclosure,Myo1p-GFPandDsRed-labeledmitochondriawerevisualizedin

30 cellsthatborelargebudsattheonsetofimagingfor2hrs.Duringthistime,contractile ringclosureoccurredin100%ofthewild-typecellsexamined(n=19)andinonly29%of the mdm10 ∆cellexamined(n=38).Toevaluatemitochondrialinheritanceasafunction ofcontractileringclosure,wemeasuredthemitochondrialcontentinbudsof mdm10 ∆ cells that undergo contractile ring closure (Fig. 2. 4 B) and in the first buds (d1) of multibudded mdm10 ∆thatfailedtoundergocontractileringclosureatthemothercell:d1 junction (Fig. 2. 4 E). In wild-type and mdm10 ∆ cells that undergo contractile ring closure 43±2% (n = 32), and 36.7±3% (n=37) of mitochondria are in the bud, respectively.Incontrast,therearenodetectablemitochondriain87%ofd1cellswithin multibudded mdm10 ∆cells(n=100).

31

Figure 2.4. Multibudded clusters of mdm10 ∆ cells are due to defects in contractile ring closure. A-D) Still frames from time-lapse imaging of Myo1p-GFP(green)andDsRed-labeled mitochondria(red)insynchronizedwild- type (ISY008) (A) and mdm10 ∆ (ISY009) (B-D) cells. Unbudded cells were isolated from mid-log phase culturesbycentrifugationthrougha10- 35%sorbitolgradientfor12minat56x g and visualized by 4D time lapse imaging 1 hr after bud formation for a totalof1hr.Imageswereacquiredat3 and 4 min intervals for wild-type and mdm10 ∆ cells, respectively. Images shown are 2D projections of 3D reconstructions. Arrows point to buds. Numbers indicate time of image acquisition from the onset of bud formation. Bar, 1 µm. A) Wild-type cell undergoing contractile ring closure. B) mdm10 ∆ cell that has mitochondria in the bud and undergoes contractile ring closure.C) mdm10 ∆cellsthatdoesnot undergocontractileringclosureandhas no detectable mitochondria in the bud. D) Multibudded mdm10 ∆ cell in which the first bud (d1) has no detectable mitochondria,andacontractileringhas assembled at the site of growth of the second daughter cell (d2). E) Mitochondrial morphology and distribution in multibudded cells from synchronized mdm10 ∆cells.Cellswere growninSCmediumat30 0Ctomid-log phase(OD 600 =0.5–0.8)andincubated with a-factor (10 µM) for 2.5 hrs. Cells were washed and resuspended in medium, fixed at various times after release from pheromone-induced G 1 arrestandstainedwithCalcufluorwhite tostainbudscarsonthemothercell(m) butnotonthefirstorseconddaughter cell (d1 and d2, respectively) produced from that mother cell (middle panel). DsRedlabeledmitochondriaarepresent in the mother cell but not in daughter cells (left panel). Bar, 1 µm. F) Quantification of mitochondrial content in mother cells (m), their first (d1) and second (d2) daughter cells in multibudded mdm10 ∆ cells from synchronized cell cultures. n = 100 clumpswith3cellbodies.

32 Role for the MEN in regulation of cell cycle progression in mdm10 ∆ cells.

The MEN regulates cell cycle progression in response to spindle alignment and elongation,andtothetransferofthenucleusfrommothertodaughtercellduringthe anaphase-to-telophase transition. Cdc14p activation and localization of the active protein to its sites of action are essential for degradation of a mitotic cyclin (Clb2p), inactivation of a mitotic cyclin-dependent kinase (CDK; Cdc28p/Clb2p), dephosphorylation of CDK substrates, and exit from mitosis (Stegmeier and Amon,

2004). However, several studies indicate that the MEN also has a direct role in regulatingcontractileringclosureduringcytokinesisinbuddingyeast(Bembeneketal.,

2005;Blondeletal.,2005;Cliffordetal.,2008;Corbettetal.,2006;Lucaetal.,2001;

SongandLee,2001).

mdm10 ∆cellsundergomitoticexit,asassessedbydegradationofClb2pandspindle disassembly(Fig2.3).Toevaluatetheroleofthe MEN in the observed cytokinesis defect, we studied the localization of Cdc14p-GFP in mdm10 ∆ and wild-type cells.

Cdc14pisreleasedfromitsinhibitorCfi1p/Net1pinthenucleolusduringtwostagesin thecelldivisioncycle.Inearlyanaphase,separase,aspartoftheCdcfourteenearly- anaphaserelease(FEAR)pathway,promotesatransient andpartialreleaseofCdc14p fromthenucleolus.Inasecond phase,signaltransductionthroughtheMENreleases the remaining Cdc14p, which facilitates mitotic exit and cytokinesis (D'Amours and

Amon,2004).

33

We confirmed that Cdc14p-GFP in wild-type cells localizes to the nucleolus through earlystagesofthecelldivisioncycle,andisreleasedfromthenucleolusandlocalizes tothespindlepolebodiesandbudneckasthespindleapparatuselongates(Fig.2. 5A).

When the spindle is at its maximum length (6-8 µm), 100% of the Cdc14p-GFP is released from the nucleolus (Fig. 2.5C). In mdm10 ∆ cells, some cytosolic Cdc14p localizes to the spindle pole body in mdm10 ∆ cells bearing fully elongated spindles.

However, release of Cdc14p-GFP from the nucleolus is inhibited by 50% in mdm10 ∆ cells bearing 4-6 µm spindles, and to a lesser extent in cells with 6-8 µm spindles comparedtowildtypecells(Fig.2.5B-C).Thus,deletionof MDM10 resultsinadelayin releaseofCdc14pfromthenucleolus.

34

Figure 2.5. Cdc14p is mislocalized in mdm10 ∆cells. Wild-type (LGY020) and mdm10 ∆ (LG0Y21) cells expressingCdc14p-GFPandmCherry-taggedtubulinwere growntomid-logphase,fixedandstainedwithDAPIasfor Fig. 2.1. The images shown are 2-D projections from reconstructed 3-D volumes. An overlay of Cdc14p-GFP (green)andtubulininthemitoticspindle(red)areshown (left).AnoverlayofCdc14p-GFP(green)andDAPI(blue) areshown(right).Celloutlinesareshowninwhite.White arrow:spindlepolebody.Whitearrowhead:nucleus.Red arrowhead:mother-budneck.Bar,1µm.A)Cdc14p-GFP localizationinwild-typecells.Cdc14p-GFPlocalizestothe nucleolus in cells bearing short, but detectable spindles (upperpanels),tothenucleusandspindlepolebodiesin early anaphase when spindles are 4-6 µm in length (middlepanels)andtospindlepolebodiesandthemother- budneckduringtelophasewhenspindleshaveelongated and reached their maximum length of 8-10 µm (lower panels). B). Defects in localization of Cdc14p-GFP in mdm10 ∆cells. C)QuantitationofthereleaseofCdc14p from the nucleolus in wild-type and mdm10 ∆ cells as a functionofspindlelength.Errorbarsshowstandarderror ofthemean(n>200).

Sli15p,achromosomalpassengerproteinandsubstrateforCdc14pthatispresentat kinetochores during metaphase and transfers to the spindle midzone during early anaphase(D'AmoursandAmon,2004),localizestothespindletothesameextentin mdm10 ∆andinwild-typecells(datanotshown).

35

Thus, mislocalization of Cdc14p in mdm10 ∆ cells is due to an alteration in MEN- mediatedcontrolofCdc14pandnottheFEARpathway.Inlightofthesefindingsand our observation that release of Cdc14p from the nucleolus is partially inhibited in mdm10 ∆ cells, it is possible that the level of MEN-mediated Cdc14p activation in mdm10 ∆cellsissufficienttosupportmitoticexitbutinsufficienttosupportcytokinesis.

Consistent with this, conditions that hyperactivate the MEN promote cytokinesis in mdm10 ∆cells.Deletionof BUB2 suppressesthesubtlemitoticexitdefectobservedin mdm10 ∆cells,buthasnoeffectonthetimeofentryof mdm10 ∆cellsintoanaphase

Deletion of BUB2 or overexpression of CDC5 in mdm10 ∆ cells results in a 67% decreaseinthenumberofmultibuddedcellsinlate-logphasecellculturescomparedto mdm10 ∆cells(Fig.2.6A-B).Thus,conditionsthatbypassMENregulationbypassthe cytokinesis defects observed in mdm10 ∆cells. To determine whether other mutations that inhibit mitochondrial inheritance also affect cytokinesis, we studied GEM1 , a member of the rho (Miro) family of GTPases and MMR1 , a protein that localizes to mitochondria, binds to the type V myosin Myo2p and is required for anchorage of mitochondriainthebudtip(Itohetal.,2002)(Fredericketal.,2008). mmr1 ∆or gem1∆ mutants exhibit subtle defects in mitochondrial inheritance, and low but detectable defects in cytokinesis. However, gem1 ∆ mmr1 ∆ double mutants exhibit mitochondrial distributionandinheritancedefectsthataresignificantlygreaterthanthoseobservedin either single mutant (Frederick et al., 2008) and a cytokinesis defect that is more

36 severethanthatobservedineithersinglemutantsandsimilartothatobservedinthe mdm10 ∆ mutant. In addition, deletion of BUB2 suppresses the cytokinesis defect observed in the gem1 ∆ mmr1 ∆ double mutant (Fig. 2.6C). These findings provide additionalevidencefortheexistenceofamechanismtoinhibitcellcycleprogressionat cytokinesiswhenthereareseveredefectsinmitochondrialinheritance.

Finally, the primary function of a checkpoint is to ensure that critical cell division processesoccurwithhighfidelityandatthecorrecttimeascellsdivide.Thus,ifthe

MEN regulates cell cycle progression in response to mitochondrial inheritance, then hyperactivation of the MEN should reduce the fidelity of mitochondrial inheritance.

Indeed, we find that conditions that bypass MEN regulation, deletion of BUB2 or overexpression of CDC5 , result in defects in partitioning of mitochondria between mother cells and buds (Fig. 2.6D). Deletion of BUB2 reduces the amount of mitochondria indaughter cells. Deletionof MDM10 producesmore severe defects in thefidelityofmitochondrialinheritance.Finally, mdm10 ∆mutantsbearingadeletionin

BUB2 or overexpressing CDC5 exhibit defects in mitochondrial partitioning that are moreseverethanthatin mdm10 ∆mutants.

37

Figure 2.6. Hyperactivation of the MEN suppresses the defect in cytokinesis defect observed in mdm10 ∆ cells. A) mdm10 ∆cellsthatexpressedmitochondria- targeted DsRed and contained either no plasmid (ISY002) or plasmid-borne CDC5 undercontroloftheGALpromoter(ISY048) incubatedingalactose-basedmediafor5.5 hrs. Images are phase-contrast images superimposed on fluorescence images of DsRed-labeledmitochondria. Bar, 3 µm. B) Quantitation of multibudded cells in wild- type cells and mdm10 ∆ cells that overexpress CDC5 or carry a BUB2 deletion. Wild-type, CDC5 overexpression, mdm10 ∆ and mdm10∆ overexpressing CDC5 strainsISY001,ISY048,ISY002and ISY013. Wild-type, bub2 ∆, mdm10 ∆, and bub2 ∆ mdm10 ∆ strains are BY4741, 6189, 398, and LGY025. Cell culture and quantitationwerecarriedoutasforFig.2.1 D. Error bars show standard deviations (n>800). C) Quantitation of multibudded cells in wild-type (BY4741), mmr1 ∆ (4139), gem1 ∆(357), mmr1 ∆gem1 ∆(DCY001)and mmr1 ∆gem1 ∆bub2 ∆cells( DCY002 )thatwereanalyzedaftersonicationandtreatment withzymolyase20T,aproteinmixturethatcatalyzescellwalldegradation,(0.1mg/ml for10minatRT).Errorbarsshowstandarddeviations( n>100).D) Hyperactivationof themitoticexitnetworkresultsinmitochondrialpartitioningdefects.Mid-logphasewild- type, bub2 ∆, mdm10 ∆, mdm10 ∆ bub2 ∆ and CDC5 overexpressing cells (ISY001, ISY028, ISY002, ISY029, and ISY013), which express mitochondria-targeted DsRed, were fixed, and images of yeast bearing large buds (buds >2/3 the length of their mothercells)werecollectedat1-µmz-intervals.Mitochondrialareainthemothercellor budwasmeasuredineachz-sectionusingauser-definedthreshold,andtheseareas were summed over the mother cell or bud to determine mitochondrial volume. Mitochondrialpartitioningratiosarethemitochondrialvolumeinthebud/mother.Error barsshowstandarderrorofthemean(n>250).

38

Overall,therearenumerouscellcyclecheckpoints tomonitor events associated with nuclear inheritance, including replication of nuclear DNA and segregation of chromosomes and nuclei. Here, we provide evidence for a mitochondrial inheritance checkpointthatinhibitscytokinesiswhentherearedefectsinmitochondrialinheritance inbuddingyeast,andforarolefortheMENinthisprocess.Sincethemitochorehas been implicated in association of mitochondria withER(Kornmannetal.,2009),itis possiblethattheseinteractionscouldcontributetocytokinesis.Moreover,in Drosophila melanogaster, mitochondrialsecondmessengers,eitherROSorATP,canfunctionas twoindependentsignalstoenforcecheckpointsatG1/Sthatarenotduetometabolic restriction (Owusu-Ansah et al., 2008). Our findings indicate that a checkpoint for mitochondrial inheritance, that is also independent to metabolic restriction, exist in buddingyeast.Finally,sincetherearemechanismstoinsuretheinheritanceofmany organellesandtheMENisaconservedpathway,ourfindingsalsoraisethepossibility thattherearesimilarcheckpointsfororganelleinheritanceinyeastandothercelltypes.

ExperimentalProcedures

Asummaryofthematerialsandmethodsusedforthisstudyisincluded.Pleasereferto

SupplementalInformation formoredetaileddescription.

Yeaststrains,plasmids,andgrowthconditions:Yeaststrainsusedinthisworkarelisted inTable2.1.StrainISY065isaderivativeofW303.StrainsMYY291andDNY416were derivedfromA364A.AllotherstrainswerederivedfromBY4741.rho0derivativeswere

39 generatedfromwild-typecellsexpressingplasmid-bornemitochondria-targetedDsRed

(ISY001),asdescribedbyGoldringetal.(Goldringetal.,1970).Otheryeastmethods wereperformedaccordingtoSherman(Sherman,2002).

ThecarboxyterminusofMyo1pandCdc14pweretaggedwithGFPusingPCR-based insertion into the chromosomal copies of the MYO1 or CDC14 loci (Longtine et al.,

1998).Table2.2listsprimersusedtotagthesegenes.Standardmoleculartechniques forcloningprocedureswereused.Mitochondriawerevisualizedusingafusionprotein expressed from the plasmid pRS426ADH + PreFoATPase-(subunit 9)-DsRed or from theplasmidpTDT104GAL1+PreFoATPase-(subunit9)-DsRed(giftsfromDr.J.Shaw,

University of Utah). Tubulin was visualized using fusion proteins expressed from the plasmid [pAFS125 TUB1-GFP] or [pRS406 TUB1-mCherry] (gifts from Dr. K. Bloom,

University of North Carolina at Chapel Hill). GAL-CDC5 bub2 ∆ cells (A4453) and plasmid pGal(myc)3CDC5-306 were gifts from Dr. A. Amon (MIT) and plasmid

[pGP195-2 (pRS305-SLI15-GFP KanMX6)] a gift from Dr. E Schiebel (University of

Heidelberg). Growth conditions for individual experiments are described in the figure legends.

To construct the mmr1 ∆ gem1 ∆ double mutant (DCY001), MMR1 was deleted in a gem1 ∆ strain (357)using an insertion cassette in which the selectable marker LEU2 wasflankedwithloxPrecombinationsites. LEU2 waslaterexcisedfromDCY001using plasmid-borne CreLox under control of the galactose inducible promoter (pSH47). To construct the mmr1 ∆ gem1 ∆ bub2 ∆ triple mutant (DCY002), pSH47, which carried a

40

URA3 marker,wascuredfromDCY001usingFOA,and BUB2 was deletedusing an insertioncassettecontaininga LEU2 marker.

Yeast cell viability was measured using FUN-1, a halogenated unsymmetric cyanine dye that was developed for assessing the viability and metabolic activity of yeast

(Millardetal.,1997).FUN-1ismembranepermeate,thatbindstonucleicacidsandis biochemicallyprocessedinlivingyeasttoproduceacylindricalintravacuolarstructure that is red shifted in fluorescence emission compared to the unprocessed form.

Incubation of yeast with FUN-1 was carried out according to manufacturer’s recommendations (Invitrogen - Molecular Probes, Carlsbad, CA). The conversion of

FUN-1byviableyeastcellswasquantifiedusingfluorescencemicroscopy.Cellswith prominent fluorescent intravacuolar structures were scored as live. Cells that lacked thesestructuresandhaddiffusecytosolicfluorescencethatwasgreenoryellowwere scoredasnon-viable.

Fluorescencemicroscopy,imageanalysisandcytology:Cellsweregentlypelletedand mounted directly in 2% low melting agarose on a coverslip. Fluorescence/phase microscopicimageswerecollectedusinganE600microscope(Plan-Apo100X/1.4NA objective) (Nikon, Melville, NY) equipped with a cooled CCD camera (Orca-ER,

Hamamatsu,Japan),andaDual-Viewimagesplitter(OpticalInsights,Tucson,AZ)for simultaneoustwo-colorimaging.Openlab3.1.5software (Improvision, Lexington, MA) wasusedtoacquireimages.Z-stacksof0.2-µmsliceswereobtainedandtheout-of-

41 focus light was removed using an iterative deconvolution algorithm in Volocity 2.6

(Improvision,Lexington,MA).Allz-sectionswereassembledand3-Dprojectionswere generatedwithcomparableparametersandthresholds.

Each cluster of more than two attached cell bodies was counted as cell separation failure and those cells were scored as multibudded cells. To stain bud scars, formaldehyde-fixed cells were incubated in 10 µg/ml Calcofluor (Invitrogen Molecular

Probes, Carlsbad CA) for 30 min at RT. For cell wall digestion, cells were fixed by incubation with formaldehyde (3.7%) for 1 hr at RT. After washes to remove the fixative, cells were incubated in 0.1 mg/ml zymolyase 20T (Seikagaku Corp., Tokyo

Japan)for10minatRT.TostainDNA,formaldehyde-fixedcellswereincubatedwith1

µg/ml DAPI (Molecular Probes, Eugene, OR) for 5 min at RT. For cell cycle synchronization, cells were incubated with α-factor (10 µM) for 2.5 hrs. Cells were released from arrest by washing and were transferred to pheromone-free media.

InductionofCDC5expressionincellscarryingthepGal(myc)3CDC5-306plasmidwas carriedoutbygrowthinSC-Raff(2%)mediumandtransfertoSCmediumcontaining raffinose(2%)andgalactose(2%).

Flowcytometry:AnalysisofDNAcontentinpropidiumiodide-stained,synchronizedcell cultures was determined according to Paulovich and Hartwell using a fluorescence- activated cell analyzer (Becton DickersonLSRII, Franklin Lakes, NJ). The percent of

42 total cells in G2 phase was determined using the Flowjo program (TreeStar Inc.,

Ashland,OR).

Proteinandimmunologicaltechniques:Proteinextractsofmid-logphaseyeastcellsfor

Western blot analysis were obtained as described (Boldogh et al., 1998). The bicinchoninic acid (BCA) assay (Pierce Chemical, Rockford, IL) was used for protein concentration determinations. Immunoblot analysis of the total amount of Clb2p and

Hxk1pwasperformedwithantibodiesspecificforClb2p(agiftfromDr.DougKellogg,

UniversityofCalifornia)andHxk1p(agiftfromDr.GottfriedSchatz,UniversityofBasel).

HRP-conjugated secondary antibodies and Supersignal detection (Pierce Chemical,

Rockford,IL)wereusedtovisualizebands.

Yeaststrains,plasmids,andgrowthconditions:

YeaststrainsusedinthisworkarelistedinTable2.1. rho 0derivativesweregenerated from wild-type cells expressing plasmid-borne mitochondria-targeted DsRed (ISY001), as described by Goldring et al. (Goldring et al., 1970). Other yeast methods were performedaccordingtoSherman(Sherman,2002).Yeastcellviability wasmeasured usingFUN-1(Millardetal.,1997).

43

ThecarboxyterminusofMyo1pandCdc14pweretaggedwithGFPusingPCR-based insertion into the chromosomal copies of the MYO1 or CDC14 loci (Longtine et al.,

1998). Standard molecular techniques for cloning procedures were used sambrook

1998Coldspringharbor.

Fluorescencemicroscopy,imageanalysisandcytology :Mitochondria,tubulinand

Sli15pwerevisualizedusingplasmidborneGFPfusionproteins.Chitininbudscarsand

DNA were visualized using CalcofluorWhite and DAPI. Acquisition, manipulation and analysis of fluorescence images was carried out as described previously (Sogo and

Yaffe,1994).

Yeaststrainsusedforthisstudy Table2.1 Strains Genotype Source

357 MAT ahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0gem1 ∆::KANMX Open Biosystems

398 MAT ahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0mdm10 ∆::KANMX Open Biosystems

4139 MAT ahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0mmr1 ∆::KANMX Open Biosystems

BY4741 MAT ahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 Open Biosystems

DCY001 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 gem1 ∆::KANMX Thisstudy mmr1 ∆::LEU2

44

DCY002 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 gem1 ∆::KANMX Thisstudy mmr1 ∆bub2 ∆::LEU2

DNY416 mdm10::URA3ura3leu2his3 Boldogh et al.,2003

ISY001 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY002 MAT amdm10 ∆:: KANMX6his3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY003 MAT amdm12 ∆:: KANMX6his3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY005 MAT a mas37∆:: KANMX6 his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY006 MAT a tom7 ∆:: KANMX6 his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY007 MAT acbk1 ∆:: HIS3his3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 Thisstudy

ISY008 MAT a MYO1-GFP :: HIS3 his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY009 MAT amdm10 ∆:: KANMX6MYO1-GFP :: HIS3 his3 ∆1leu2 ∆0 Thisstudy

met15 ∆0ura3 ∆0[pRS426ADH+PreF 0ATPase-DsRed]

ISY013 MAT a mdm10 ∆:: KANMX6 CDC14-GFP :: HIS3 his3 ∆1 Thisstudy leu2 ∆0 met15 ∆0 ura3 ∆0 [pGal(myc)3CDC5-306]

[pTDT104GAL1+PreF 0ATPase-DsRed]

ISY016 MAT a CDC14-GFP :: HIS3 his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY018 MAT a mdm10 ∆:: KANMX6 CDC14-GFP :: HIS3 his3 ∆1 Thisstudy

leu2 ∆0 met15 ∆0 ura3∆0 [pRS426ADH+PreF 0ATPase- DsRed]

ISY028 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 bub2 ∆::HIS3 Thisstudy

[pRS426ADH+PreF 0ATPase-DsRed]

ISY029 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 bub2 ∆::HIS3 Thisstudy

45

mdm10 ∆::KANMX [pRS426ADH+PreF 0ATPase-DsRed]

ISY048 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 [pGal(myc)3CDC5- Thisstudy

306][pRS426ADH+PreF 0ATPase-DsRed]

ISY065 MAT ∆mmm1-1,leu2-∆1trp1-∆1his3-∆200ura3ade2his3 Thisstudy

leu2lys2trp2ura3 [pRS426ADH+PreF 0ATPase-DsRed]

LGY020 MAT a CDC14-GFP :: HIS3 his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 Thisstudy [pRS406 TUB1 -mCherry]

LGY021 MAT a mdm10 :: KANMX6CDC14-GFP :: HIS3 his3 ∆1leu2 ∆0 Thisstudy met15 ∆0ura3 ∆0 [pRS406 TUB1 -mCherry]

LGY022 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 [pRS406 TUB1 - Thisstudy mCherry][pGP195-2(pRS305-SLI15-GFP KanMX6)]

LGY023 MAT a mdm10 ∆:: KANMX6his3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 Thisstudy [pRS406 TUB1 -mCherry] [pGP195-2 (pRS305-SLI15-GFP KanMX6)]

LGY024 MAT ahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0bub2 ∆::HIS3 Thisstudy

LGY025 MAT a his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 bub2 ∆::HIS3 Thisstudy mdm10 ∆::KANMX

MYY291 ura3leu2his3 Yaffe and Smith,1991

46

CHAPTER3

MtDNAINHERITANCECHECKPOINT

47

BACKGROUND

Mitochondria contain>1,000proteins.Themajority of these proteins are encoded by nuclearDNA,undergoMendelianinheritanceandareimportedfromthecytoplasminto the mitochondria (Chen and Butow, 2005). In addition, mitochondria contain DNA

(mtDNA),whichencodesrespiratorychaincomponentsorthatareessentialfor mitochondrialproteinsynthesis.mtDNAvariesinsizeamongorganisms.Thebudding yeast Saccharomycescerevisiae hasalargemitochondrialgenomeofaround75-80kb.

In contrast, the human mitochondrial genome is 16.5 kb (Chen and Butow, 2005).

Human mtDNA contains 37 genes, 13 encode proteins that participate in oxidative phosphorylation,22encodetRNAsand2encoderibosomalRNAs(Wallace,2010).

mtDNA is organized and packaged as a DNA-protein complex called the mtDNA . Haploid cells of the yeast Saccharomyces cerevisiae with wild-type mitochondrialgenomes(rho +cells)contain10–20mtDNAnucleoidspercell(Williamson andFennell,1979).Thismacromolecularcomplexisinheritedfrommothertodaughter cells during cell division (Williamson and Fennell, 1979) (Stevens, 1981/coldspring harbor). Many proteins that are required for stabilizing and or packaging of mtDNA nucleoidshavebeenidentifiedincludingDNAbindingproteins(e.g.theabundanthigh mobilitygroup-box,mtDNA-bindingnucleoidpackingproteinAbf2p),mtDNAreplication proteins (e.g. human DNA polymerase γ and Mip1 in yeast), and transcription (e.g. humanTFAM(transcriptionfactorAandyeastAbf2)(Zelenaya-Troitskayaetal.,1998).

Another protein is Mgm101p, a DNA-binding protein that is essential for mtDNA

48 maintenance and required for repairing oxidative damaged mtDNA and localizes to a

‘subset’ area around mtDNA nucleoids (Meeusen and Nunnari, 2003) (Chen et al.,

1993).Mgm101palsointeractswithMmm1p,amitochore protein that is required for mitochondrialinheritanceandformaintenanceofmtDNA(MeeusenandNunnari,2003).

One other protein that localizes to mtDNA nucleoids and is relevant to my thesis researchisPif1.Pif1pispartofthesuperfamily1DNAhelicases,proteinsthat unwind

DNA and are essential for DNA replication, recombination, and repair (Budd et al.,

2006) (Changetal.,2009).TherearetwoformsofPif1.Onelocalizestothenucleus and contains a nuclear localization sequence. Recent studies indicate that DNA damage results in Rad53-dependent phosphorylation of Pif1 at nuclear DNA damage breaks, which inhibits telomerase activity at those sites, and prevents telomerase- dependentadditionoftelomeresatDNAbreaks(MakovetsandBlackburn,2009).The other form of Pif1 contains a mitochondrial targeting sequence, localizes to mtDNA nucleoidsandisrequiredformtDNAmaintenance(FouryandKolodynski,1983).Cheng etal.reportedthat pif1 ∆cellshavemtDNAbreaksatspecificsitesandproposedthat

Pif1peitherpreventsorrepairsmtDNAdsDNAbreaks(Chengetal.,2007).

OTHERPROTEINSIMPLICATEDINmtDNAINHERITANCE

A number of proteins on the mitochondrial surface, including the mitochore and

Mdm34p,havealsobeenimplicatedinmaintenanceofmtDNA.Theseproteinslocalize topunctatestructuresonmitochondriathatareadjacenttomtDNAnucleoidsandare

49 required for maintenance of mtDNA (Boldogh et al., 2003) (Hobbs et al., 2001)

(Youngmanetal.,2004).Thereisalsoevidencethatthemitochoremayinteractwiththe mtDNAreplicationproteincomplexes.Mip1pisasubunitofmtDNApolymerasegamma ofyeast.TheMgm101p/Mip1pproteincomplexassociateswithmtDNAnucleoidsandis essential for mtDNA replication and mtDNA genome maintenance (Meeusen and

Nunnari, 2003). Mitochore-containing foci are adjacent to and move with a protein complex consisting of Mgm101p and Mip1p. Consistent with this, Mgm101p has physicalandgeneticinteractionswiththemitochoresubunitMmm1p.Thesedatareveal a physical interaction between the mitochore and mtDNA nucleoids, and provide evidencethatthemechanismformitochondrialmotilityduringinheritanceislinkedtothe processofmtDNAreplicationandinheritance.

mtDNAMUTATIONS:rho 0ANDrho -CELLS

mtDNA has a very high mutation rate, resulting in clinical manifestations (Wallace,

2010).EpidemiologicalstudiesindicatethatthefrequencyofhumanmtDNAmutations isintheorderof1/5000(Wallace,2010).Further,geneexpressionmicroarraystudies revealedthatlossofhumanmtDNAresultsindown-regulationofcellcycleregulatory genesandareductionofcellreplicationratesresultinginalongerS-phase(Mineriet al.,2009).

Sinceyeastarefacultativeaerobes,theycanwithstandmtDNAmutationsthatarenot readilytoleratedbymammaliancells.InyeasttherearetwobroadclassesofmtDNA mutants: rho - and rho 0 cells. rho 0 cells contain no mtDNA, while rho - cells contain

50 mtDNAthatcarriesmutationsthatblockmitochondrialrespiratoryactivity.Asaresult, bothofthesecelltypeshavedefectsinrespirationandcannotgrowinmediacontaining non-fermentable carbon sources like glycerol or lactate. Moreover, upon growth on fermentable carbon sources like glucose, rho 0 and rho - cells produce small colonies calledpetites.

Earlystudiesonrho -cellsthatcarrylargedeletionsinmtDNArevealedthatyeastcells exercise tight control of the mtDNA genome size. Interestingly, the total size of the mtDNAgenomeinthese rho -cellsissimilartothatofrho +cells( ~80kb)(Dujon,1981- coldspringharborpublication).Laterstudiesrevealedthatrho -cellsmaintainthesizeof theirmtDNAgenomebyamplifyingexistingmtDNAandcreatingtandemrepeatsofthe amplifiedDNAuntilthecriticalsizeof~80kbisreached(Figure3.1).rho - cellscontain fewer mtDNA nucleoids per cell that stain more brightly with DNA binding dyes compared to rho + cells, but they still continue to contain the same mass of mtDNA

(Dujon,1981-coldspringharborpublication).

TheabilityofyeasttoamplifymtDNAtoacriticalmasssupportstheexistenceof surveillancemachinerytomonitormtDNAmass,andregulatorymachinerytoup- regulatemtDNAsynthesisuntilitreachesacriticalgenomicmass.Weidentifieda mtDNAmasssurveillancesystem,whichinhibitscellcycleprogressionfromG1toS phaseinresponsetolossofmtDNA.Moreover,wefindthatthismtDNAinheritance checkpointisregulatedbyaconservedG1toScheckpointpathway,theRad53DNA damagecheckpoint.

51

FIGURE3.1-mtDNAcontentisundertightcellularcontrol. Yeasthavetheabilityto amplybytandemrepeatssmallfragmentsofmtDNAuntilthetotalsizeofthemtDNAis similartothatfoundincellsthatcontainwildtypemtDNA(75kb).Thiscontrolsuggests thatthereisaregulatorysystemthatmonitorsacriticalmassofmtDNA.Thiscartoon depictsasmallfragmentthatisamplifiedbytandemrepeatstothecriticalsizeof75kb. Thisnew‘amplified’genomeisarespiratoryincompetentrho -cell.

Rad53ANDTHEDNADAMAGECHECKPOINT

The yeast proteins kinases, Mec1 and Rad53 are components of a conserved checkpointthatarreststhecellcycleatG1,S,andG2phaseandactivatespathwaysto repair damaged DNA in response to damage of nuclear DNA (Elledge, 1996). The responsetoDNAdamageisdependentonwhentheinsulttakesplaceandthetypeof damage that occurs. Two major checkpoints occur in response to DNA damage or replication errors. The DNA damage checkpoint functions throughout the entire cell

52 cycleincludingS-phasewhereastheDNAreplicationcheckpointpathwayisrestricted tocellsundergoingactivereplication(Schwartzetal.,2003).

Mec1isahomologofthehumanATR(Ataxia-andRad-related)gene,whileRad53isa homologofhumanCHK2(Chabesetal.,2003).InresponsetoDNAdamage,Mec1and

Ddc2 complexes are recruited to the site of damage or stalled DNA replication fork, whichleadstophosphorylationandactivationofRad53(Cordon-Preciadoetal.,2006).

ActivationofRad53leadstocellcyclearrestandactivationofgenesthatcontributeto

DNArepair.Rad53triggersaG1arrestbystimulating the phosphorylation of Swi6, whichleadstodown-regulationofthetranscriptionoftheG1cyclins, CLN1 and CLN2

(SidorovaandBreeden,1997).

Rad53 has 16 potential phosphorylation sites, a catalytic domain and two forkhead homology-associated (FHA) domains (FHA-1, FHA-2) (Lee et al., 2003). Both FHA domainsarerequiredfortheDNAdamagedependentpathway;however,eitherFHA1 orFHA2domainallowsactivationofRad53inresponsetoareplicationblock(Schwartz etal.,2003).FHAdomainsinteractwithbothupstreamanddownstreameffectorsofthe

DNA checkpoint signaling pathways (Schwartz et al., 2003). In response to DNA damage, Rad9 is phosphorylated and binds to Rad53 via the FHA-1 and FHA-2 domains.ReplicativestressactivatesRad53byaRad9 independent mechanism that mayinvolveanintermediatekinase,Mrc1(Alcasabasetal.,2001).

53

Figure 3.2 Rad53 activation during DNA damage. Checkpoints govern many responses to DNA damage and blocks to DNA replication. A. Rad9 or POL2 class genes encode proteins that recognize either damage (* denotes damage by single- strand gaps, stalled replication fork) and then activate Mec1, Rad53 ). The signal transducers mediate multiple responses and are dependent on the type of insult or where in the cell cycle the damage took place. B. Dun1 can also be activated from doublestrandedbreaksorotherDNAinsults.C.ReplicationforkrecognizeDNAlesions duetodamageordNTPdeprivation.Helicasesandpolymerasesuncoupleexposingthe DNAwhereRPAcanbindtoandactivatethecheckpointresponse.Mec1isrecruited whichinitiatesthesignalingcascadeinvolvingRad53andotherdownstreamkinases.

Ribonucleotidereductase(RNR)isanimportanttargetthatisactivatedbyRad53and servestopromoterepairofDNAdamage.RNRcatalyzestherate-limitingstepindNTP synthesis (Zhao et al., 1998) (Reichard, 1988) and affects both the fidelity of DNA replicationandcellviabilitybyregulatingthedNTPpools(ZhaoandRothstein,2002).

Rad53increasesdNTPlevelsbyactivatingthedownstreamkinaseDun1,whichleads tothetranscriptionalinductionofthe RNR genes.Inconjunction,Mec1/Rad53increase

54

RNR levels by regulating the degradation of the negative inhibitor Sml1 (Zhao and

Rothstein, 2002). Interesting, regulation of dNTP levels is the essential function of

Rad53: the Mec1 or Rad53 lethality can be suppressed by overexpression of RNR genesorbyinhibitionofSml1(SuppressorofMec1Lethality)(KocandMerrill,2007)

(Zhaoetal.,1998)(Chabesetal.,2003).

After successful repair of DNA damage, Rad53 is downregulated by phosphatase- mediated de-phosphorylation and not degradation (O'Neill et al., 2007), which allows cellstoresetcellcycleprogression(Cordon-Preciadoetal.,2006;Pelliciolietal.,2001).

DephosphorylationofRad53by PTC2 and PTC3 phosphatasescanalsoallowcellsto bereleasedfromthecheckpointwhenDNArepairattemptsfail.Duringthisprocess, whichisreferredtoasrecoveryfromthecheckpoint, cells reenter the cell cycle with damagedDNA.

LINKSBETWEENRad53ANDmtDNA

Cells contain multiple copies of mtDNA, ranging from20–50inyeasttoasmanyas

10,000 in mammalian cells. Nuclear genes have direct interactions with genes that control mtDNA replication, maintenance and copy number in mammalian cells and yeast(Tayloretal.,2005)(Tyynismaaetal.,2004)(Matsushimaetal.,2004)(Schultzet al., 1998) (Zelenaya-Troitskaya et al., 1998). While conserved genes have been implicated in this process, signaling pathways involved in modulating cellular mtDNA contenthavenotbeenfullyelucidatedinanyorganism.

55

TheShadellaboratoryobtainedthefirstevidencethatmtDNAcopynumberisregulated byDNAdamagecheckpointpathwaysinyeastandmammaliancells.Theyfoundthat the Ataxia-Telangiectasia mutated (ATM) gene, a kinase that regulates cell cycle progressioninresponsetoDNAdamage,anditstarget,ribonucleotidereductase(RNR) regulatesmtDNAcopynumberinhumancells(Eatonet al., 2007). Specifically, they found that mtDNA copy number is reduced in Ataxia-telangiectasia (A-T) patient fibroblasts,wild-typefibroblaststreatedwithanATMinhibitor,andcellsinwhichRRis inhibitedbydrugtreatmentorRNAinterference(RNAi).

Their studies in yeast revealed that deletion of the RNR inhibitor SML1 increases mtDNA copy numberandthat deletion of RAD53 and SML1 results in an increase in mtDNAcopynumberthatisgreaterthanthatobservedin sml1 ∆cells(Lebedevaand

Shadel, 2007). Thus, mtDNA copy number in yeast is regulated by Rad53, in part througheffectsonRNRactivityanddNTPpoolsize.Finally,otherstudiesindicatethat

Pif1 and RRM3, helicases that are associated with nuclear DNA and mtDNA have genetic interactions with Rad53: 1) overexpression of the RAD53 target, RNR, suppressesthelossofmtDNAassociatedwithdeletionof PIF1 ,and2)deletionof PIF1 and RRM3 resultsinactivationofRad53(Tayloretal.,2005).

Together, these studies revealed a functional link between the DNA damage checkpointsandmtDNA.Moreover,theymayexplainsomeofthesymptomsofA-Tthat are not readily explained by damage to nuclear DNA. Here, Ipresentevidencefor a mtDNA inheritance checkpoint, a pathway that inhibits progression from G1 to S in

56 responsetolossofmtDNA.Furthermore,wefindthatthecheckpointisregulatedby

Rad53,anestablishedcheckpointsignalingmoleculethathasknowneffectonnuclear

DNAandmtDNA.

RESULTS

LOSSOFmtDNAINDUCESAG1ARRESTINCELLCYCLEPROGRESSION

Treatment with ethidium bromide (EtBr) results in the conversion of respiratory competentyeast(grandes)torespiratorydeficientmutants(petites)atefficienciesclose to 100%. Early studies revealed that EtBr treatment inhibits mtDNA synthesis and increases mtDNA degradation, which results in production of rho 0 cells that have no mtDNA(Goldringetal.,1970).WeusedEtBrtogeneraterho 0cells,andconfirmedthe loss of mtDNA, by staining with the DNA binding dye 4',6-diamidino-2-phenylindole

(DAPI) which is a fluorescent stain that binds strongly to A-T rich regions in DNA.

PunctatemtDNAnucleoidsarepresentinrho +cellsthathaveintactmtDNAbutnotin rho 0 cells that have no mtDNA (Figure 3.3A). We also confirmed that loss of mtDNA results in loss of mitochondrial respiratory activity by assessing growth on a non- fermentablecarbonsource,suchasglycerol(SFig3.1).

We used flow cytometry to assess the effect of the loss of mtDNA on cell cycle progression.Wild-typerho +cellsexhibitnormalcellcycleprogression(Fig.3.3).They undergotransitionfromG1toSphase40-60min,andtransitionfromStoG2phase

57

100-120minafterreleasefrompheromone-inducedG1arrest.Incontrast,wefindthat

30-60%ofrho 0cellsfailtoprogressfromG1toSphase.Rho 0cellsthatdoprogress throughthecellcycledosoatalowerrate.They undergo transition from G1 to S phase 60-100 min, and transition from S to G2 phase 150 min after release from pheromone-inducedG1arrest.

Figure3.3.LossofmtDNAresultsindefectsinprogressionfromG1toSphase. Wild-type(BY4741)orcellslackingmtDNA(rho 0, mgm101 ∆cells)weregrowntomid- logphase(OD600=0.5–0.8)andarrestedG1phasebytreatmentwithpheromone( α- factor;10-100µm)for2.5-3hrs.Sampleswerereleased from pheromone-induced G1 arrestbywashing,resuspendinginsyntheticcomplete(SC)media,andpropagatedat 30°C. Aliquots were removed at the time shown. Cells were fixed, stained with propidiumiodine(PI)andtheirDNAcontentasafunctionoftimeafterreleasefromG1 arrest was analyzed by flow cytometry. (A) Maximum projections of deconvolved imagesofWT,rho 0and mgm101 ∆rho 0cellsstainedwiththeDNAbindingdyeDAPI.n: nuclearDNA,m:mtDNA.CelloutlinesareshowninwhiteBar,1µm.(B)Wild-typecells withmtDNA(BY4741rho +)exhibitnormalcellcycleprogression.CellslackingmtDNA either by EtBr treatment (BY4741 rho 0) or by deletion of a gene required for mtDNA maintenance ( mgm101 ∆ rho 0) fail to progress normally and show a G1 defect. (C) QuantitationofprogressionthroughG1phasewasassessedastheratioofthepercent of cells in G2 phaseat thetime of releasefrom pheromone-induced G1 arrest to the percentofcellsinG2atthetimespecified.

58

There are significantly fewer mechanisms for repair of DNA damage in mitochondria compared to nuclei. As a result, mtDNA is 10-times more sensitive to damage by chemicalinductioncomparedtonuclei(Chengetal.,2007).Therefore,itislikelythat

EtBrisaffectingmtDNAandnotnuclearDNA.However,itisformallypossiblethatthe

EtBrthatwasusedtogeneraterho 0cellsisdamagingnuclearDNAandtriggeringacell cycle progression defect through checkpoints that detect nuclear DNA damage. To addressthisissue,wegeneratedrho 0cellsbydeletionof MGM101 ,whichencodesa

DNA-bindingproteinthatisessentialformtDNAmaintenanceandrequiredforrepairof oxidatively damaged mtDNA (Meeusen and Nunnari, 2003) (Chen et al., 1993). We observed the rho 0 mgm101 ∆ cells exhibit a stronger defect in the ability to exit G1 comparedtorho 0cellsproducedbyEtBrtreatment(Fig.3.3).Thestrongerdefectincell cycleprogressioninrho 0mgm101 ∆cellsmaybearesultoftherapidlossofmtDNAthat occurs upon deletion of MGM101 . Together, this data indicates that loss of mtDNA resultsindefectsincellcycleprogressionfromG1toSphase.

THEG1TOSPROGRESSIONDEFECTOBSERVEDINCELLSLACKINGmtDNAIS NOTDUETOLOSSOFMITOCHONDRIALRESPIRATORYACTIVITYORENERGY PRODUCTION

Previousstudiesrevealedthelossofmitochondrialrespiratoryactivityresultsinadefect inprogressionfromG1toSphasein Drosophila (Mandaletal.,2005) (Owusu-Ansahet al.,2008).Thesestudiesrevealedthatdefectsin mitochondrial function produced by eitherdeletionofcytochromeoxidasecomplexVaordisruptionofcomplex1resultsin

59 tworetrogradesignalsthatleadtodown-regulationofcyclinEandROSsignaling.To determinewhethertheG1toSphasetransitiondefectobservedinrho 0cellsisdueto loss of mitochondrial energy production, we assessed cell cycle progression in cells treated with oligomycin, an agent that binds to the F 1F0 ATPase proton pump and inhibits ATP production and respiration-driven yeast cell growth on non-fermentable carbonsources(Fig.3.4).Flowcytometryrevealedthattreatmentwitholigomyocinhad noeffectoncellcycleprogression.Thus,defectsintheG1toSphaseprogressionin rho 0cellsisnotduetolossofmitochondrialATPproduction.

FIGURE3.4-InhibitionofATPproductiondoesnotcausethedefectinpassage throughG1incells. (A)Effectofoligomycintreatmentongrowthofwildtyperho+cells on fermentable and non-fermentable carbon sources. Cells were propagated in rich mediacontainingglucose,glycerolorglycerolandethanolinthepresenceorabsence of oligomycin (1 µg/mL) shown for 48 hours, and cell density was assessed by determiningtheopticaldensityofthecultureat600nm.(B)Cellcycleprogressionof wild-type mtDNA-containing cells in the absence or presence of oligomycin. Cell synchronization, propagation in glucose based media and analysis by flow cytometry was carried out as for Fig 3.3. (C) Quantitation of progression into G2 phase was carriedoutasforFig.3.3.

60

Next, we assessed the effect of deletion of subunit 5a of cytochrome c oxidase

(COX5A )oncellcycleprogression.First,weconfirmedthatdeletionof COX5A results indefectsinrespirationdrivenyeastcellgrowth (datanotshown). Moreover,wefound thatcellcycleprogressionof cox5a ∆rho +cellsissimilartothatobservedinwild-type rho +cells(Fig.3.5).Finally,wefoundthatdeletionofmtDNAin cox5a ∆cells( cox5a ∆ rho 0)resultsindefectsinG1toSphasetransitionthatissimilarthatobservedinrho 0 cells(Fig.3.5).TheseresultsconfirmthefindingthatlossofmtDNAresultsinG1toS phase transition defects, and that the observed defects are not due to loss of mitochondrialrespiratoryactivity.

Figure3.5-Lossofmitochondrialrespiratoryactivitydoesnotcausethedefectin passagethroughG1. (A)Maximumprojectionsofdeconvolvedimagesof cox5a ∆rho + and cox5a ∆ rho 0cellsstainedwiththeDNAbindingdyeDAPI.n:nuclear DNA, m: mtDNA.CelloutlinesareshowninwhiteBar,1µm.B)Cellcycleprogressionof cox5A ∆ cellswithorwithoutmtDNA( cox5a ∆rho+, cox5a ∆rho 0respectively)weredetermined asforFig.3.1.ImagesofDAPIstainedcellsare2Dprojectionsofthereconstructed3D volume.(C)QuantitationofprogressionthroughG2phasewascarriedoutasforFig. 3.3.

61

THEDEFECTINCELLCYCLEPROGRESSIONOBSERVEDINrho 0CELLSISDUE TOLOSSOFDNAINMITOCHONDRIAANDNOTGENESENCODEDBYTHATDNA

To determine whether the observed cell cycle delay is due to loss of DNA and not genesencodedbymtDNA,westudiedcellcycleprogressioninacellthathasmtDNA butdoesnothaveanygenesencodedbythatmtDNA. ThemtDNAofthecellused containsatandemrepeatsofa71kbfragmentofthecytochromebgene(N24rho -)

(Tzagoloffetal.,1979).Thisrho -cellcontainsmtDNAnucleoids(Fig.3.6A);however, sincethemtDNAhasnogenes,thecellisrespiratory incompetent and cannot grow usinganon-fermentablecarbonsource (Datanotshown).

Figure3.6LossofmtDNAfunctiondoesnotactivate a cellular delay . Rho +, rho - andrho 0cellsweregrowntomid-logphaseinglucose-basedmedia(OD600=0.5–0.8). Unbuddedcellswereisolatedbycentrifugationthrougha10%–35%sorbitolgradientfor 12minat56xg. UnbuddedcellswerepropagatedinfreshSCmedia,andcellcycle progression was analyzed as for Fig. 3.3. (A) Images of DAPI stained cells are 2D projectionsofthereconstructed3Dvolume.(B)Cells with wild-type mtDNA (rho +) or mutantDNAwithnogenes(N24rho -)havenormalcellcycleprogression.Cellslacking mtDNA (rho 0) exhibit defects in G1 to S progression. (C) Quantitation of cell cycle progressionwascarriedoutasforFig.3.3.

62

WeassessedcellcycleprogressionintheN24rho -cellandinrho +andrho 0cellsinthe samegeneticbackgroundastheN24rho -cell(D273-10b)(Fig.3.6).LossofmtDNAin theD273-10bgeneticbackgroundalsoresultsinadefectinG1toSphasetransition.

Thus, the observed cell cycle progression defect is not a consequence of genetic backgroundused.Equallyimportant,wefoundthattheN24rho -cellhasasimilarcell cycleprogression profiletothatseeninthewild-typerho +cell.Thus,thedefectinG1to

Sphasetransitionobservedinrho 0cellsisduetolossofDNAwithinmitochondriaand notduetolossofgenesencodedbymtDNA.

ROLEFORAKNOWNCHECKPOINTPROTEIN(Rad53)INREGULATIONOFCELL CYCLEPROGRESSIONINCELLSLACKINGmtDNA

Rad53isaconservedcellcycleregulatoryproteinthatcanarrestthecellcycleatthe

G1 to S phase transition, and has functional interactions with mtDNA in mammalian cellsandyeast.Thisraisesthepossibilitythatthecellcycledefectsobserveduponloss of mtDNA may be regulated by Rad53. To test this hypothesis, we tested whether deletionof RAD53 willsuppressthecellcycleprogressiondefectsobservedinrho 0cells

63

Figure3.7Deletionof SML1 ,aninhibitorofdNTPsynthesis,doesnotsuppress the cell cycle progression defect observed in upon loss of mtDNA Cell cycle progressionofwild-type(W303),sm1 ∆rho+orEtBrderivedsml1 ∆rho 0 wascarriedout asdescribedinFig.3.3.(A)ImagesofDAPIstainedcells.n:nuclearDNA,m:mtDNA. Bar:1µm.(B-C)CellcycleprogressionwasassessedasforFig.3.3.

Since RAD53 is an essential gene and deletion of SML1 suppresses the lethality of rad53 ∆,weassessedtheeffectofdeletionofmtDNAin rad53 ∆sml1 ∆and sml1 ∆cells.

Deletionof SML1 doesnotsuppressthedefectinG1toStransitionobserveduponloss of mtDNA (Fig. 3.7). In contrast, cell cycle progression of rad53 ∆/sml ∆ rho 0 cells is similartothatobservedin rad53 ∆/sml ∆rho + cells(Fig.3.8).Thefindingthatdeletionof

Rad53allowsforcellcycleprogressioninarho 0cellprovidesadditionalsupportforthe findingthatrho 0exhibitadefectinprogressionfromG1toSphase.Italsosupportsa roleforRad53inregulationofcellcycleprogressioninresponsetolossofmtDNA.

64

Figure3.8RoleforRad53inregulatingcellcycleprogressioninresponsetoloss ofmtDNA (A-C)Deletionof RAD53 suppressesthecellcycledefectobservedinrho0 cells.Cellcycleprogressionofrho +andrho 0rad53 ∆/sml1 ∆cellswasassessedasfor Fig.3.3.(A)ImagesofDAPIstainedcells.n:nuclearDNA,m:mtDNA.Bar:1µm.(B-C) CellcycleprogressionwasassessedasforFig.3.3.(D)Pif1,aRad53targetiselevated inrho 0cellscomparedtorho +cells.Rho +andrho 0cellscontainingPif1pthatistagged at its chromosomal locus with 13 copies of the myc epitope were grown to mid-log phase in glucose based rich media. Cells were concentrated by centrifugation, and extracted with SDS sample buffer. Proteins in the whole cell extracts were analyzed usingWesternblotsandantibodiesraisedagainstthemycepitopeandhexokinase.The levelsofhexokinaserevealedthatequalamountsofproteinwereanalyzedinrho +and rho 0cells.Usingthemycantibody,itisclearthatmyc-taggedPif1pispresentathigher levelsinrho 0comparedtorho +cells.

IfRad53isregulatingcellcycleprogressionattheG1toSphasetransitioninresponse tolossofmtDNAthenlossofmtDNAshouldresultinactivationofRad53p.Totestthis, wecomparedthesteadystatelevelofPif1p,aDNAhelicasethatisrequiredformtDNA maintenanceandhasgeneticinteractionswithRad53,inrho +andrho 0cells.Wefound

65 thatthesteadystatelevelsofPif1pareelevatedby45%inrho 0cellscomparedtorho +

(Fig.3.8D).ThisresultindicatesthatdeletionofmtDNAresultsinactivationofRad53p.

DISCUSSION

Surveillance of nuclear DNA to ensure genomic integrity before the replication and segregation to a new daughter cell has been studied for several decades. The involvementoftheATR/Mec1anddownstreamkinasessuchasRad53hasbeenwell accepted throughout the field. Here, we report that the existence of a surveillance systemthatinhibitscellcycleprogressionfromG1toSphasewhenthereisalossof mtDNA.

In Drosophila , defects in mitochondrial respiratory activity triggers a checkpoint that inhibitsG1toSprogression(Mandaletal.,2005;Owusu-Ansahetal.,2008).However, we find that treatment with an agent that inhibits mitochondrial ATP production or deletion of a mitochondrial respiratory chain component, which results in loss of respiratoryactivity,doesnotaffecttransitionfromG1toSphaseinyeast.Instead,we findthattheobservedcellcycledefectistriggeredbylossofDNAwithinmitochondria andnotbylossofthegenesencodedbymtDNA.Specifically,wefindthatrho -yeast, whichcontainmtDNAthatissimilarinsizetowildtypemtDNAbutdoesnotbearany genes,exhibitswildtypeprogressionfromG1toSphase.

Furthermore, we have implicated a known checkpoint signaling pathway, the Rad53

DNADamagecheckpoint,incontrolofprogressionthroughG1inresponsetolossof

66 mtDNA. Previous studies indicate that deletion of RAD53 results in an increase in mtDNAcopynumberinyeastandmammaliancells,andthatPif1p,aDNAhelicasethat localizestonucleiandmitochondria,hasgeneticinteractionswithRad53andisatarget forRad53. Thus,thereisanestablishedfunctionallinkbetweentheRad53pathwayand mtDNA.WefindthatPif1pisup-regulatedinresponsetolossofmtDNA.Consistent withthis,wefindthatdeletionof RAD53 by-passestheG1toStransitiondefectincells withnomtDNA.

Although the essential function of Rad53 is in regulation of dNTP pools, our studies indicatetheregulationofdNTPpoolsizealoneisnotresponsibleforthedefectsinG1 toSprogressionincellswithoutmtDNA.Specifically,wefindthatdeletionof SML1 ,a negativeregulatoroftheenzymethatcatalyzestherate-limitingstepindNTPsynthesis

(Rnr1p), does not suppress the G1 to S transition defect observed in cells without mtDNA.Thus,ourevidencesupportsthemodelthatRad53functionsinregulatingcell cycleprogressioninresponsetolossofmtDNAasacheckpointsignalingmoleculeand notasaregulatorofdNTPlevels.

Our previous studies revealed a checkpoint that inhibits cytokinesis in response to defects in inheritance of mitochondria and is regulated by the Mitotic Exit Network

(Chapter 2). This checkpoint is triggered by loss of mitochondria and not by loss of mtDNA. Here, we report the identification of a second checkpoint that monitors mitochondrialinheritance.Inthiscase,thetriggerforcellcycledelayislossofDNAin mitochondria.Accordingly, werefertothischeckpointasthemtDNAcheckpoint.Our

67 dataalsosupportaroleforRad53inregulationofthemtDNAinheritancecheckpoint.

Thus,wefindthatRad53checkpointfunctionisbroaderthanpreviouslyappreciated:it monitorsDNAdefectsinboththenucleusandmitochondria. Finally, since Rad53 is conserved,andaffectmtDNAinyeastandmammaliancells,itispossiblethatamtDNA checkpointexistsincellsotherthanyeast.

SupplementalFig3.1(SFig3.1)Growthofallstrainsusedinthisstudyonfermentable andnon-fermentablecarbonsources.Atitrationseriesofstrainonglucose-based media(YPD)andglycerol-basedmedia(YPG).Rho0andcellsbearingadeletioninthe COX5genehavedefectsinmitochondrialrespiratoryactivityandcangrowona fermentablecarbonsource(glucose)butnotonanon-fermentablecarbonsource (glycerol).

68

Defectsinrespirationcanbereadilydetectedbyassayingforgrowthonanon- fermentablecarbonsource.Allviableyeaststrainexhibitgrowthonmediacontaining fermentablecarbonsources.However,growthonnon-fermentablecarbonsources requiresmitochondrialrespiratoryactivity.Asaresult,rho 0cellsgrowonglucosebutnot onglycerolbasedmedia.Allrho 0hadthepredictedphenotype:LackofDAPInucleoid stainingandglycerolgrowth(supplementalFig3.1).

EXPERIMENTALPROCEDURES

Yeaststrains,plasmids,andgrowthconditions:

YeaststrainsusedinthisworkarelistedinTable3.2.StrainISY065isaderivativeof

W303.StrainsMYY291andDNY416werederivedfromA364A. rho 0derivativeswere generated from wild-type by two consecutive two day treatments of 25 µM EtBr

(Goldring et al., 1970). Standard molecular techniques for cloning procedures were used sambrook 1998 Coldspring harbor(28). Other yeast methods were performed accordingtoSherman(Sherman,2002).

Tagging of Pif1: The carboxy terminus of PIF1 was tagged with 13-myc using PCR- basedinsertionintothechromosomalcopiesofthepif1loci(27).Primersusedtotag these genes were: forward primer

CGAACCTCGTGGTCAGGATACCGAAGACCACATCTTAGAACGGATCCCCGGGTTA

ATTAA and reverse primer

GCAGTTTGTATTCTATATAACTATGTGTATTAATATGTACGAATTCGAGCTCGTTTAAAC.

69

Standardmoleculartechniquesforcloningprocedureswereused.Growthconditionsfor individualexperimentsaredescribedinthefigurelegends.

Synchronization: Forcellcyclesynchronization,cellswereincubatedwith α-factor(10-

100µM)for2.5hrs.Cellswerereleasedfromarrestbywashingandweretransferredto pheromone-freemedia.

Flow cytometry: Analysis of DNA content in propidium iodide-stained, synchronized cell cultures was determined according to Paulovich and Hartwell (Paulovich and

Hartwell,1995)usingafluorescence-activatedcellanalyzer(BectonDickersonLSRII,

FranklinLakes,NJ).ThepercentoftotalcellsinG1phasewasdeterminedusingthe

Flowjoprogram(TreeStarInc.,Ashland,OR).

Fluorescencemicroscopy,imageanalysisandcytology: Cellsweregentlypelleted, resuspended in fresh media and mounted directly onto a pad consisting of 2% low melting agarose and SC media. The sample was covered with a coverslip and fluorescence/phase microscopic images were collected using an E600 microscope

(Plan-Apo 100X/1.4 NA objective) (Nikon, Melville, NY) equipped with a cooled CCD camera (Orca-ER, Hamamatsu, Japan), and a Dual-View image splitter (Optical

Insights, Tucson, AZ) for simultaneous two-color imaging. Openlab 3.1.5 software

(Improvision, Lexington, MA) was used to acquire images. Z-stacks of 0.2-µm slices wereobtainedandtheout-of-focuslightwasremovedusinganiterativedeconvolution

70 algorithm in Volocity 5.5 (Improvision, Lexington, MA). All z-sections were assembled and3-Dprojectionsweregeneratedwithcomparableparametersandthresholds.

Proteinandimmunologicaltechniques: Proteinextractsofmid-logphaseyeastcells for Western blot analysis were obtained as described (Boldogh et al., 1998). The bicinchoninic acid (BCA) assay (Pierce Chemical, Rockford, IL) was used for protein concentration determinations. Immunoblot analysis ofthetotalamountof(Pif1)was performedwithantibodiesspecificformyc.HRP-conjugatedsecondaryantibodiesand

Supersignaldetection(PierceChemical,Rockford,IL)wereusedtovisualizebands.

71

Table3.1.

Strains Genotype Source

BY4741 MATahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0 OpenBiosystems DCY023 MATahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0rho 0 Thisstudy 6937 MATahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0mgm101 ∆ OpenBiosystems 7210 MATahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0cox5a ∆ OpenBiosystems DCY033 MATahis3 ∆1leu2 ∆0met15 ∆0ura3 ∆0cox5a ∆rho 0 Thisstudy W1588-4C MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 R.Rothstein his3-11,15 DCY025 MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 Thisstudy his3-11,15rho 0 U952-3B MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 R.Rothstein kan-his3-11,15sml1 ∆::HIS3 DCY027 MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 Thisstudy kan-his3-11,15sml1 ∆rho 0 U960-5C MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 R.Rothstein his3-11,15sml1-1rad53 ∆::HIS3 DCY029 MAT a leu2-3,112 trp1-1 can1-100 ura3-1 ade2-1 Thisstudy his3-11,15sml1-1rad53 ∆::HIS3rho 0 D273-10B MAT αmal G.Schatz DCY015 MAT αmalrho 0 Thisstudy N24 MAT αmalN24rho- Nobrega and Tzagoloff, 1980 PDY001 MATa his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 PIF1-13- Thisstudy myc PDY002 MATa his3 ∆1 leu2 ∆0 met15 ∆0 ura3 ∆0 PIF1-13- Thisstudy mycrho 0

72

CHAPTER4

DISCUSSION

73

DISCUSSION

Therearemanysurveillancemechanismsthatcontroltheprogressionofthemitoticcell cycleinresponsetonuclearinheritance.Recentfindingsrevealthatthesesurveillance mechanismsarenotlimitedtomonitoringtheinheritance of nuclei, but thatthey can alsosenseandrespondtothedistributionandinheritanceofotherorganellesincluding mitochondria, cER and Golgi. The finding that checkpoint surveillance occurs for organellesthatcanandcannotbeproduceddenovo underscores the importance of organelle inheritance to cell division and provides additional evidence that organelle inheritance is an integral component of the cell division cycle. It also raises the possibility that there are organelle inheritance checkpoints for other organelles in the cell including and peroxisomes. Indeed, emerging evidence supports the existenceofcheckpointsforinheritanceofthe()inyeast(Barelleet al.,2003;Vesesetal.,2009).

Recentfindingsalsoindicatethatthecheckpointsforinheritanceofmitochondria,cER andGolgi are regulated by conserved cellcycle regulatory mechanisms.This reveals novel functions for known pathways and suggests that mechanisms underlying organelle inheritance checkpoints are also conserved. Yet to be determined is the mechanism(s) for communication between checkpoint machineries and their target organelle.Whatisthemechanismfordetectingdefectsininheritanceofmitochondria and other organelles? How is information from inheritance checkpoint sensors

74 transmittedtothemachinerythatmediatescellcyclearrest?Doorganellecheckpoint pathwaysactivateproteinsthatpromoteorrepairorganelleinheritancewhenthereare defectsintheseprocesses?Ifso,how?Futurestudieswillprovideanswerstoeachof thesefundamentalquestionsonthecheckpointmachineriesofmitochondriaandother organelles.

SUMMARYOFPROJECT1:

Myfirstprojectrevealedthattheinheritanceofmitochondriainthebudisrequiredfor cellcycleprogressionatcytokinesis.Wepublishedthatmutationsthatresultinsevere defects in mitochondrial inheritance also results in defects in cell cycle progression throughcytokinesis.Themutantsusedinthesestudiesareyeastbearingadeletionof

MDM10 , which results in defects in mitochondrial movement during inheritance, or deletion of MMR1 ,aproteinthatmediatesanchorageofmitochondriainthebudtip, combinedwithdeletionof GEM1 ,acalciumbindingGTPasethatisrequiredfornormal mitochondrialdistribution. Deletionof MDM10 inhibitscontractileringclosure,butdoes notinhibitcontractileringassembly,localizationofachromosomalpassengerproteinto thespindleduringearlyanaphase,spindlealignment,nucleolarsegregationornuclear migration during anaphase. Release of the mitotic exit network (MEN) component,

Cdc14p,fromthenucleolusduringanaphaseisdelayedin mdm10 ∆cells.

We also showed that this mitochondrial inheritance checkpoint is regulated by the mitoticexitnetwork(MEN).Ifoundthatthemitochondrialinheritancecheckpointcould bebypassedinboththe mdm10 ∆and mmr1 ∆gem1 ∆cellsbyhyper-activatingtheMEN

75 networkbyeitherknockingoutthenegativeinhibitor BUB2 or by overexpressing the polokinaseCdc5p.Finally,sincecheckpointsaredesignedtoincreasethefidelityof processesthatarecriticalforthecelldivisioncycle,wetestedwhetherinhibitionofthe

MEN compromised mitochondrial inheritance. Here, we found that deletion of BUB2 resultsinadecreaseinthefidelityofmitochondrial segregation between mother and daughtercellsinwild-typeand mdm10 ∆cells.Overall,ourstudiesidentifyanovelMEN- linkedregulatorysystemthatinhibitscytokinesisinresponsetodefectsinmitochondrial inheritanceinbuddingyeast.

FUTUREEXPERIMENTS:

An important goal for future studies is to identify the sensor that detects defects in mitochondria inheritance, and how this sensor communicates that information to the

MEN.Preliminarydataobtainedfrommystudiesandstudiesfromothermembersofour laboratoryserveasafoundationforthesestudies.

Ifoundthatthepassageofonlyasmallfragmentofmitochondriaacrossthebudneck caninitiatecontractileringclosureandcommittocytokinesis.Preliminaryexperiments supportaroleforDbf2andMob1inthisprocess.Dbf2andMob1arepartofaser/thr kinase complex that is involved in transcription and stress responses. The complex functions as part of the MEN pathway and is activated by the MEN kinase Cdc15p.

Dbf2p has kinase activity and Mob1p binds to Dbf2, which results in conformational changes in Dbf2p that allows an upstreamkinase, Cdc15p, to phosphorylate Mob1p

76 andDbf2p.PhosphorylationofDbf2pactivatesitskinaseactivity,whichisrequiredfor cytokinesisandcellseparation.

Duringearlystagesinthecelldivisioncycleinwild-typecells,Mob1pandDbf2plocalize tothecytosol.Duringanaphase,Mob1pandDbf2prelocalizetothespindlepolebody andmotherbudneck,wheretheyactivateproteinsthat contribute to mitotic exit and cytokinesis.OngoingstudiesindicatethatMob1pand Dbf2p are mislocalized in two strains( mdm10 ∆and mmr1 ∆gem1 ∆)withseveredefectsinmitochondrialinheritance andcytokinesis.Bothproteinsaredetectedinthespindlepolebodyandatthebud neckinmitochondrialinheritancemutants.However,Mob1pandDbf2palsolocalizeto mitochondria in mdm10 ∆ and mmr1 ∆ gem1 ∆ mutants. This raises the possibility that

Mob1pandDbf2parepartofthesensorfordetectingmitochondriainheritanceduring the yeast cell division cycle and that they do so through direct interactions with mitochondria.Moreover,sinceMob1pandDbf2areconservedinallcellsstudied,itis possiblethattheymayserveassensorsformitochondrialinheritanceinothercelltypes.

Thesefindingsraiseadditionalquestions:whatproteins do Mob1p and Dbf2 interact withonmitochondria?HowisbindingofMob1pandDbf2ptomitochondriaregulated?

DoesmislocalizationofDbf2pandMob1ptomitochondriainmitochondrialinheritance mutantspreventtheactivecomplexfromlocalizingtoitssiteofactionatthemother-bud neck?

77 SUMMARYOFPROJECT2:

MysecondprojectshedslightonthesurveillancemechanismformtDNAandhowthe presenceofmtDNAisrequiredfornormalG1toStransitionduringyeastcelldivision.

Manyotherlaboratorieshavepreviouslypublishedtherepairmechanismneededwhen thereisdamage,defectsinreplication,ordefectsinthesegregationofnuclearDNA; however,wearethefirsttoshowthatthereisasurveillancemechanismfordefectsin mtDNA.ForDNAdamagecheckpoints,thedamagedetectedisnotassociatedwitha specificgeneorsetofgenes.Rather,thesecheckpointsmonitorbroaderprocesses, includingstalledDNAreplicationfork,singleordoublestrandedDNAbreaks.Wehave observedsimilarfindingsasinthemtDNAcheckpoint.ThemtDNAcheckpointobserved inyeastalsodoesnotmonitorspecificgenesorfunctionofgenesencodedbymtDNA.

Instead,itmonitorsmtDNAcontent.

My results also indicate that the loss of mtDNA activates a DNA damage signaling pathway, Rad53, which results in a defect in progression from G1 to S phase.

Specifically,IfoundthatthelossofmtDNAresultsinanincreaseinthesteadystate levelsofPif1p,aproteinthatisactivatedbytheDNAdamagecheckpointandthoughtto be a substrate of Rad53. This finding indicates that Rad53 is activated by loss of mtDNA.Moreover,sincePif1pisaDNAhelicasethatlocalizesbothtothenucleusand themitochondrion,thisfindingindicatesthatactivated Rad53p also promotes mtDNA repairinresponsetolossofmtDNA.Consistentwiththis,IfoundthatdeletionofRad53

78 by-passes the cell cycle progression defect observed in cells that do not contain mtDNA.

Although regulation of dNTP pools is an essential function of Rad53, my studies suggestthatthearrestfoundinacelllackingmtDNA(rho 0)isnotduetothedecreasein deoxynucleotides (dNTP). Specifically, I found that deletion of the ribonucleotide inhibitor (RNR) SML1, whichincreasesdNTPpools,didnotrestorenormal cell cycle progressioninacellthathasnomtDNA.Instead, my data supports the model that

Rad53 functions in the mtDNA inheritance checkpoint as a regulator of G1 to S transition,andnotasaregulatorofdNTPlevels.

Our findings are supported by reports in the literature in which cell cycle arrest was observedincellsthathavelosttheirmtDNA.Oneofthesestudiesfocusedon DNA2 ,an essentialgenethatencodesaendonuclease,and5'to3'DNAhelicasethatcontributes toOkazakiFragmentprocessingandrepairofdoublestrandDNAbreaks.Mutationof

DNA2 resultsinacellcyclearrestthatismediatedbytheMec1/Rad53pathway.(Budd etal.,2006).Interestingly,mutationsof DNA2 resultsinahighrateofmtDNAloss,and cells that have lost their mtDNA undergo cell cycle arrest (Fiorentino and Crabtree,

1997). Thus,otherlaboratorieshaveobservedalinkbetweenmtDNAloss,cellcycle progressiondefectsandtheRad53DNAdamagepathway.

79

ROLEFORDNAPol γγγASASENSORFORTHEmtDNAINHERITANCE

CHECKPOINT:

TheonlyDNApolymerasefoundsolelyinanimalcellmitochondriaisDNApolymerase gamma(POLG).ThispolymerasebearsthesoleresponsibilityforDNAsynthesisinall replication, recombination, and repair transactions involving mitochondrial DNA

(mtDNA).Humanpolymerase γiscomposedofa140kDacatalyticsubunitA(POL γA) and a 55 kDa accessory subunit B, which increases the processivity of mtDNA synthesis.POL γAcomprisesapolymerasedomainandanexonucleaseproofreading domain,separatedbyalinkerregionof482aminoacids(Figure4.1).

Over60PEO-associatedmutationshavebeenfoundinPOL γγγ(Leeetal.,2009)(Viikov et al., 2011). Most of the dominant POL γγγ mutations are in the polymerase domain, whereas most of the recessive mutations are in the exonuclease or in the spacer domains. Mutations in Pol γγγ are associated with a spectrum of disease phenotypes including autosomal dominant and recessive forms of progressive external ophthalmoplegia,(PEO),spino-cerebellarataxiaandepilepsy,andAlpers-Huttenlocher hepatocerebral poliodystrophy (Lee et al., 2009). Multiple deletions, or depletion of mtDNAinaffectedtissues,arethemolecularhallmarksofpol γγγmutations.

80

Figure 4.1 Taken from (Lee et al., 2009) to illustrate the various domains of human polymerasegamma.

TheyeastorthologueofPOL γγγis MIP1 ,whichencodesforthecatalyticsubunitofDNA polymerase γγγ.Proteomicandgeneticscreenshaverevealedthat Mip1, like Abf2and

Mgm101, is associated with mtDNA nucleoids (Meeusen and Nunnari, 2003). Mip1 directly binds to mtDNA, synthesizes DNA stretches of up to several thousand nucleotides without dissociation from the template, carries out DNA synthesis on

81 double-stranded templates utilizing a strand displacement mechanism, and is active evenincellsthathavenomtDNA(Viikovetal.,2011).

DNA repair complexes that associate with DNA at site of damage influence the checkpoint response by facilitating the recruitment of checkpoint factors or by generatingtheintermediateDNAstructuresthatfunctionassignalstothecheckpoint machinery.AlthoughtherearelimitedrepairpathwaysformtDNA,theonlyproteinthat functionsinthetwoknownrepairmechanisms,singlenucleotidebaseexcisionrepair andlongpatchbaseexcisionrepair,isMip1p.Moreover,unlikeothermtDNAbinding proteinslikePif1pandAbf2pthatarepresentinreducedlevelsinmitochondriawithno mtDNA,Mip1localizestomitochondriaandisfullyfunctionalinrho 0cells(Viikovetal.,

2011). This raises the possibility that Mip1p may serve as a sensor for the mtDNA inheritancecheckpoint.

IfMip1pisamtDNAinheritancecheckpointsensor, then deletion of MIP1 should by- passtheG1toStransitiondefectobservedinrho 0cells.Indeed,Ifoundthatdeletionof

MIP1bypassesthecellcycledelayassociatedwithlossofmtDNAinrho 0cellsthatare madethroughthedeletionof MGM101 (Figure4.2A). Thus,preliminarydataindicate that MIP1 ispartofthesensorthatdetectsmtDNAandactivatesRad53inresponseto loss of mtDNA. Interestingly, Mip1p may function as a sensor for defects in mtDNA replicationbecauseitislocatedatthereplicationfork.However,itwouldbeimpossible

82 todistinguishbetweenasensoryroleversusasignaltransductionroleanditispossible thatitistheactivityofanentirecomplexandnotasinglepolymerasethatmustbeintact toproperlysensereplication(Elledge,1996).

Figure 4.2 Role for DNA polymerase gamma, Mip1, in the mtDNA inheritance checkpoint-Cellprogressionisnotaffectedbyknockingoutmgm101inmip1 ∆cells. (A)Cellscycleprogressionofmip1 ∆and mip1 ∆mgm101 ∆rho 0cells.(B)Quantitation ofcellcycleprogressionin mip1 ∆and mip1 ∆mgm101 ∆rho 0withcontrols(WTRho+ andmgm101 ∆cells)wasdeterminedasdescribedinFig3.2.

TofurtherelucidatethepossibilitythatMip1isamtDNAinheritancesensor,Iwouldtake advantageofpreviousworkbyBaruffini,E.Lodi,T.etal.andotherswhostudiedMIP1 mutations associated with mitochondrial diseases in humans (Baruffini et al., 2006).

Specifically, I would be interested in studying the mutation within the exonuclease domain(G303R)thathasaseverephenotypeandcompletedepletion of mtDNA and

83 compare it to DNA binding mutations (R574W) and (P625R) that results in in less severephenotypes(Baruffinietal.,2006;Leeetal.,2009).Iwouldpredictthatthecell cycle regulation in response to loss of mtDNA may be dependent upon Mip1 DNA bindingbutnotnecessarilydependentuponDNApolymeraseactivity.

This approach may also be used to identify other proteins that regulate the mtDNA inheritancecheckpoint.Here,Iwouldscreenfor MIP1 mutationsthatinhibititsfunction inthemtDNAinheritancecheckpoint,butdonotaffectitsfunctioninmtDNAreplication.

Thereafter,Iwouldperforma2-hybridscreentoidentifyproteinsthatinteractwithWT

MIP1 but do not interact with MIP that is defective in its mtDNA sensor function.

Finally,IwouldtestiftheseproteinsinteractwithMIP1invivo,andwhetherdeletionof candidateMip1-interactingmtDNAsensorproteinssuppressthecellcycleprogression defectsobservedinrho 0cells.

HOW IS THE SIGNAL THAT mtDNA LOSS TRANSMITTED FROM MITOCHONDRIATOTHENUCLEUS?

Themitochondrialretrograde(RTG)pathwayisapathwayfor communicationbetween the mitochondria and the nucleus. This pathway is active in both normal and pathologicalconditionsandinvolvesmultiplefactorsthatsenseandtransmitsignalsto effectchangesinnucleargeneexpression.Thesechangesleadtoareconfigurationof metabolism to accommodate cells with mitochondrial defects (Liu and Butow, 2006).

Retrograde signaling has been connected to nutrient sensing, TOR signaling, growth

84 control, and other signaling processes that function in metabolic and organelle homeostasis,aswellasaging(LiuandButow,2006).

Interestingly,theRTGpathwayaffectsmtDNAmaintenance through regulation of the

RTG target gene, ACO1 (Liu and Butow, 2006). ACO1 is one of many bifunctional genesinmitochondria.Aco1p,aconitase,functionasametabolicenzymeintheTCA cycleandasasignalingmoleculeintheRTGpathway.EarlierstudieslinkedAco1to mtDNA inheritance through the identification of Aco1p being among proteins cross- linkedtomtDNA.Equallyimportant,thelossofmtDNAobservedupondeletionofthe mtDNApackagingproteinAbf2pcanbesuppressedbyactivatingACO1expressionand the RTG pathway (Liu and Butow, 2006). Moreover, Aco1p localizes to mtDNA, and deletionofACO1resultsinseveremtDNAinstability(LiuandButow,2006).Sinceloss ofmtDNAisnotobservedupondeletionofotherTCAcyclegenes,mtDNAinstability observedinACO1mutantsisattributetoACO1functionintheRTGpathwayandnot centralmetabolism.(McCammonetal.,2003).Thesestudiesleadtotheconclusionthat aconitasehastwodistinctfunctionandactivities: 1) enzymatic activity and 2) mtDNA maintenanceactivity.

In light of this, it would be important to test whether the RTG pathway serves as a signalingpathwaytoactivateRad53inresponsetolossofmtDNA.Todoso,Iwould testwhethermutationofACO1orotherRTGsignalingmoleculesby-passesthemtDNA

85 inheritance checkpoint. Specifically, I would study G1 to S cell cycle progression,

Rad53phosphorylationandPif1plevelsinrho 0cellsandrho 0cellsbearingmutationsin

ACO1 and other RTG signaling molecules. Given evidence for a role of the RTG pathwayinthemtDNAinheritancecheckpoint,IwouldbeginthesearchforRTGtargets thatplayaroleinthisprocessbyanalysisofgenesthatareup-regulatedinresponseto activationofRTGthathavegeneticorphysicalinteractionswithRad53.

POSSIBLE ROLE FOR PROHIBITINS IN THE mtDNA INHERITANCE CHECKPOINT

Prohibitinwasoriginallyidentifiedasagenethat inhibits cell cycle progression when microinjectedintomouseembryonicfibroblasts(MEFs)(Osmanetal.,2009a).Inyeast there are two known prohibitin genes, prohibitin-1 (PHB1) and prohibitin-2 (PHB2), which share more than 50% identical amino acid residues, are conserved and are present in all eukaryotes sequenced to date (Van Aken et al., 2010). Moreover, prohibitins localize and function predominantly to the mitochondria and the mitochondrial inner membrane and are involved in mitochondria fusion through the regulation of the dynamin-like GTPase OPA1 (Osman et al., 2009b) (Merkwirth and

Langer,2009).

Interestingly, prohibitins localize to different cellular compartments, such as, the nucleus, the plasma membrane, and mitochondria. In fact PHB2 has been shown to

86 translocatefromthemitochondriatothenucleusfollowing the binding of estradiol by

ER α (Osman et al., 2009b). This raises the possibility that prohibitins function in signalingfrommitochondriatothenucleus,inadditiontotheirfunctioninregulationof mitochondrialfusion.Consistentwiththis,mouseembryonicfibroblasts(MEFs)lacking

PHB2 were partially rescued by overexpressing the non-cleavable L-OPA1 isoform

(Merkwirthetal.,2008).

Equally important, prohibitins function in the maintenance and stability of mtDNA.

Crosslinking studies revealed that PHB1 and PHB2 are peripheral components of mtDNA nucleoids (Wang and Bogenhagen, 2006) (Bogenhagen et al., 2008).

Furthermore,Kasashimaetal.(2008)observedalinkbetweenprohibitinsandsteady state of nucleoids. They found that downregulating PHB1 expression in HeLa cells resultedinadecreaseinthelevelsofTFAMandmtDNA.Theabsenceofprohibitinalso leads to an increased generation of reactive oxygen species, disorganized mtDNA nucleoids, abnormal cristae morphology, inhibition of complex I of the mitochondrial electrontransportchainandanincreasedsensitivitytowardsstimuli-elicitedapoptosis

(Osmanetal.,2009b)(Artal-SanzandTavernarakis,2009).Thesefindingssuggestthat

PHB1 maintains the organization and copy number of mtDNA by regulating TFAM stability (Osman et al., 2009b) (Kasashima et al., 2008). An important goal of future studiesistodeterminewhetherprohibitinsserveassignalingmoleculesinthemtDNA inheritancecheckpoint.

87 HOWMYSTUDIESMAYCONTRIBUTETOOURUNDERSTANDINGOF MITOCHONDRIALDISEASES

Therehasbeensomecontroversyontheinterpretationbetweenhumansamplesandin vitro cellular studies in regards to mitochondrial diseases. Human patients with mitochondrialdiseasehaveadeteriorationoftherespiratorychainfunctionandthrough randomsegregationeventsmayincreaseordecreasethemutatedmtDNA.Impairment inmitochondrialrespiratoryfunctionnotonlyreducesthesupplyofenergy,whichmay prevent energy-dependent apoptosis, but also enhances ROS production that may induce mutation and oxidative damage to mitochondrial DNA (mtDNA) (Lee et al.,

2005). The accumulation of mtDNA mutations and alteration leading to increase apoptosis contributes to the onset and progression of various neurodegenerative diseases(Leeetal.,2005).Recently,itwasreportedthattheexpressionlevelofthe β subunitofF1-ATPaserequiredformitochondrialATPsynthesisisdecreasedinhuman cancers,includingliver,gastric,lung,andcolorectalcancers(Leeetal.,2005).

These types of results calls into question the proposed quality control mechanisms makingitclearthatdifferentcellularmodelneedstobeconstructed.Amutatormouse hasbeenmadebyTrifunovicetal.thatshowsastrongphenotype,interestinglynotat embryonic or early development but at 25 weeks (Trifunovic et al., 2004). This premature aging phenotype and model is caused by the accumulation of point mutationsandthepresenceoflineardeletedmtDNAmolecules(Trifunovicetal.,2004)

(ParkandLarsson,2011).

88

Decreasedmitochondrialoxidativephosphorylation(OXPHOS)isoneofthehallmarks of cancer. Mutations in POLG are known to cause mtDNA depletion and decreased

OXPHOS,resultinginmtDNAdepletionsyndromeinhumans.AstudybySinghetal. revealedthatmtDNAwasdepletedinbreasttumors.Consistently,mutantPOLG,when expressed in breast cancer cells, induced a depletion of mtDNA, decreased mitochondrial activity, decreased mitochondrial membrane potential, and increased levelsofreactiveoxygenspecies.HefurtherproposedthatdecreasedOXPHOSledto anincreasedrateofaerobicglycolysisinmostcancers.Thisphenomenonisdescribed astheWarburgeffect(Singhetal.,2009).

LaboratorieshavemeasuredmtDNAcontentdirectlyintumorsandreportadecreasein mtDNA content in breast, renal, hepatocellular, gastricandprostatetumors(Singhet al.,2009).Itisalsonoteworthythatdrugsusedfor treating human immunodeficiency virus (HIV) inhibit POLG, which in turn induces mtDNA depletion (Kakuda, 2000).

Tamoxifen, a commonly used drug for the treatment of breast cancer, also depletes mtDNA (Larosche et al., 2007). A recent study also demonstrates that depletion of mtDNAcorrelateswithtumorprogressionandprognosisinbreastcancerpatients(Yuet al.,2007).

Manydiseaseshavebeenlinkedtoamutationwithinacheckpointpathway.Themost understood pathway studied in mamamalian cells is the p53 DNA damage pathway.

89

Thep53geneisatumorsuppressorthatismutatedinmanyhumancancercellsand encodesatranscriptionfactorthatisactivatedinresponsetoDNAdamageandleadsto increasednucleotidepools.Cellsthataredefectiveinthep53genecannotinduceaG1 arrestandshowsareducedabilitytoinduceanapoptoticresponse.Checkpointssuch asp53arethetargetsforchemotherapeutictreatment.

Therefore,itispossiblethatamtDNAinheritancecheckpointexistsinmammaliancells, andthatdefectsinthisprocessmaycontributetocancer.Myprojectidentifiedfastand simple readouts to identify proteins and the signaling pathways involved in the regulationduringsegregationofbothmitochondriaanditsgenome,andtousebudding yeastasatranslationaltooltostudyhumandisease.Itisalsoimportanttonotethat there is no, as of yet, transfection system for mammalian mitochondria, and mouse modelseitherintroduceanaturallyoccurringmtDNAmutationintomouseembryosor manipulatenucleargenesthatcontrolmaintenanceandexpressionofmtDNA.These limitations and the fact that yeast can propagate without the need to respire or use oxidativephosphorylationtosurvivemakesitanidealmodelsystemforthesehuman disease studies. Thus, my research can be used to identify and further understand signalingpathwaysandfunctionbetweenthemitochondria, its genome,and how it is regulated during cell division in yeast and in mammalian cells including those that contributetohumandiseases.

90

BIBLIOGRAPHY

91

Alcasabas, A.A., A.J. Osborn, J. Bachant, F.H. Hu, P.J.H. Werler, K. Bousset, K. Furuya, J.F.X. Diffley, A.M.

Carr, and S.J. Elledge. 2001. Mrc1 transduces signals of DNA replication stress to activate Rad53. Nat Cell

Biol . 3:958-965.

Altmann, K., M. Frank, D. Neumann, S. Jakobs, and B. Westermann. 2008. The class V myosin motor protein, Myo2, plays a major role in mitochondrial motility in Saccharomyces cerevisiae. J Cell Biol . 181:119-130. Artal-Sanz, M., and N. Tavernarakis. 2009. Prohibitin and mitochondrial biology. Trends Endocrinol Metab . 20:394-401. Barelle, C.J., E.A. Bohula, S.J. Kron, D. Wessels, D.R. Soll, A. Schafer, A.J. Brown, and N.A. Gow. 2003. Asynchronous and asymmetric vacuolar inheritance in true hyphae of Candida albicans. Eukaryot Cell . 2:398-410. Baruffini, E., T. Lodi, C. Dallabona, A. Puglisi, M. Zeviani, and I. Ferrero. 2006. Genetic and chemical rescue of the Saccharomyces cerevisiae phenotype induced by mitochondrial DNA polymerase mutations associated with progressive external ophthalmoplegia in humans. Hum Mol Genet . 15:2846-2855. Bembenek, J., J. Kang, C. Kurischko, B. Li, J.R. Raab, K.D. Belanger, F.C. Luca, and H. Yu. 2005. Crm1- mediated nuclear export of Cdc14 is required for the completion of cytokinesis in budding yeast. Cell Cycle . 4:961-971. Blondel, M., S. Bach, S. Bamps, J. Dobbelaere, P. Wiget, C. Longaretti, Y. Barral, L. Meijer, and M. Peter. 2005. Degradation of Hof1 by SCF(Grr1) is important for actomyosin contraction during cytokinesis in yeast. EMBO J . 24:1440-1452. Bogenhagen, D.F., D. Rousseau, and S. Burke. 2008. The layered structure of human mitochondrial DNA nucleoids. J Biol Chem . 283:3665-3675. Boldogh, I., N. Vojtov, S. Karmon, and L.A. Pon. 1998. Interaction between mitochondria and the actin cytoskeleton in budding yeast requires two integral mitochondrial outer membrane proteins, Mmm1p and Mdm10p. J Cell Biol . 141:1371-1381. Boldogh, I.R., D.W. Nowakowski, H.C. Yang, H. Chung, S. Karmon, P. Royes, and L.A. Pon. 2003. A protein complex containing Mdm10p, Mdm12p, and Mmm1p links mitochondrial membranes and DNA to the cytoskeleton-based segregation machinery. Mol Biol Cell . 14:4618-4627. Boldogh, I.R., and L.A. Pon. 2007. Mitochondria on the move. Trends Cell Biol . 17:502-510. Boldogh, I.R., H.C. Yang, W.D. Nowakowski, S.L. Karmon, L.G. Hays, J.R. Yates, 3rd, and L.A. Pon. 2001. Arp2/3 complex and actin dynamics are required for actin-based mitochondrial motility in yeast. Proc Natl Acad Sci U S A . 98:3162-3167. Broustas, C.G., and H.B. Lieberman. 2011. Contributions of RAD9 to tumorigenesis. J Cell Biochem . Budd, M.E., C.C. Reis, S. Smith, K. Myung, and J.L. Campbell. 2006. Evidence suggesting that Pif1 helicase functions in DNA replication with the Dna2 helicase/nuclease and DNA polymerase delta. Mol Cell Biol . 26:2490-2500. Campellone, K.G., and M.D. Welch. 2010. A nucleator arms race: cellular control of actin assembly. Nat Rev Mol Cell Biol . 11:237-251. Chabes, A., B. Georgieva, V. Domkin, X. Zhao, R. Rothstein, and L. Thelander. 2003. Survival of DNA damage in yeast directly depends on increased dNTP levels allowed by relaxed feedback inhibition of ribonucleotide reductase. Cell . 112:391-401. Chang, M., B. Luke, C. Kraft, Z. Li, M. Peter, J. Lingner, and R. Rothstein. 2009. Telomerase is essential to alleviate pif1-induced replication stress at telomeres. Genetics . 183:779-791.

92

Chen, X.J., and R.A. Butow. 2005. The organization and inheritance of the mitochondrial genome. Nat Rev Genet . 6:815-825. Chen, X.J., M.X. Guan, and G.D. Clark-Walker. 1993. MGM101, a nuclear gene involved in maintenance of the mitochondrial genome in Saccharomyces cerevisiae. Nucleic Acids Res . 21:3473-3477. Cheng, X., S. Dunaway, and A.S. Ivessa. 2007. The role of Pif1p, a DNA helicase in Saccharomyces cerevisiae, in maintaining mitochondrial DNA. Mitochondrion . 7:211-222. Clifford, D.M., B.A. Wolfe, R.H. Roberts-Galbraith, W.H. McDonald, J.R. Yates, 3rd, and K.L. Gould. 2008. The Clp1/Cdc14 phosphatase contributes to the robustness of cytokinesis by association with anillin-related Mid1. J Cell Biol . 181:79-88. Corbett, M., Y. Xiong, J.R. Boyne, D.J. Wright, E. Munro, and C. Price. 2006. IQGAP and mitotic exit network (MEN) proteins are required for cytokinesis and re-polarization of the actin cytoskeleton in the budding yeast, Saccharomyces cerevisiae. Eur J Cell Biol . 85:1201-1215. Cordon-Preciado, V., S. Ufano, and A. Bueno. 2006. Limiting amounts of budding yeast Rad53 S-phase checkpoint activity results in increased resistance to DNA alkylation damage. Nucleic Acids Res . 34:5852-5862. D'Amours, D., and A. Amon. 2004. At the interface between signaling and executing anaphase--Cdc14 and the FEAR network. Genes Dev . 18:2581-2595. Dai, Y., and S. Grant. 2011. Methods to study cancer therapeutic drugs that target cell cycle checkpoints. Methods Mol Biol . 782:257-304. Dimmer, K.S., S. Jakobs, F. Vogel, K. Altmann, and B. Westermann. 2005. Mdm31 and Mdm32 are inner membrane proteins required for maintenance of mitochondrial shape and stability of mitochondrial DNA nucleoids in yeast. J Cell Biol . 168:103-115. Eaton, J.S., Z.P. Lin, A.C. Sartorelli, N.D. Bonawitz, and G.S. Shadel. 2007. Ataxia-telangiectasia mutated kinase regulates ribonucleotide reductase and mitochondrial homeostasis. J Clin Invest . 117:2723-2734. Elledge, S.J. 1996. Cell cycle checkpoints: preventing an identity crisis. Science . 274:1664-1672. Fagarasanu, A., M. Fagarasanu, and R.A. Rachubinski. 2007. Maintaining populations: a story of division and inheritance. Annu Rev Cell Dev Biol . 23:321-344. Fagarasanu, A., and R.A. Rachubinski. 2007. Orchestrating organelle inheritance in Saccharomyces cerevisiae. Curr Opin Microbiol . 10:528-538. Fehrenbacher, K.L., I.R. Boldogh, and L.A. Pon. 2005. A role for Jsn1p in recruiting the Arp2/3 complex to mitochondria in budding yeast. Mol Biol Cell . 16:5094-5102. Fehrenbacher, K.L., H.C. Yang, A.C. Gay, T.M. Huckaba, and L.A. Pon. 2004. Live cell imaging of mitochondrial movement along actin cables in budding yeast. Curr Biol . 14:1996-2004. Fiorentino, D.F., and G.R. Crabtree. 1997. Characterization of Saccharomyces cerevisiae dna2 mutants suggests a role for the helicase late in S phase. Mol Biol Cell . 8:2519-2537. Foury, F., and J. Kolodynski. 1983. pif mutation blocks recombination between mitochondrial rho+ and rho- genomes having tandemly arrayed repeat units in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A . 80:5345-5349. Frederick, R.L., K. Okamoto, and J.M. Shaw. 2008. Multiple pathways influence mitochondrial inheritance in budding yeast. Genetics . 178:825-837. Frenz, L.M., S.E. Lee, D. Fesquet, and L.H. Johnston. 2000. The budding yeast Dbf2 protein kinase localises to the centrosome and moves to the bud neck in late mitosis. J Cell Sci . 113 Pt 19:3399- 3408. Garcia-Rodriguez, L.J., D.G. Crider, A.C. Gay, I.J. Salanueva, I.R. Boldogh, and L.A. Pon. 2009. Mitochondrial inheritance is required for MEN-regulated cytokinesis in budding yeast. Curr Biol . 19:1730-1735.

93

Garcia-Rodriguez, L.J., A.C. Gay, and L.A. Pon. 2007. Puf3p, a Pumilio family RNA binding protein, localizes to mitochondria and regulates mitochondrial biogenesis and motility in budding yeast. J Cell Biol . 176:197-207. Goldring, E.S., L.I. Grossman, D. Krupnick, D.R. Cryer, and J. Marmur. 1970. The petite mutation in yeast. Loss of mitochondrial deoxyribonucleic acid during induction of petites with ethidium bromide. J Mol Biol . 52:323-335. Hartwell, L.H., J. Culotti, and B. Reid. 1970. Genetic control of the cell-division cycle in yeast. I. Detection of mutants. Proc Natl Acad Sci U S A . 66:352-359. Hartwell, L.H., and M.B. Kastan. 1994. Cell cycle control and cancer. Science . 266:1821-1828. Hartwell, L.H., and T.A. Weinert. 1989. Checkpoints: controls that ensure the order of cell cycle events. Science . 246:629-634. Hobbs, A.E., M. Srinivasan, J.M. McCaffery, and R.E. Jensen. 2001. Mmm1p, a mitochondrial outer membrane protein, is connected to mitochondrial DNA (mtDNA) nucleoids and required for mtDNA stability. J Cell Biol . 152:401-410. Honlinger, A., U. Bomer, A. Alconada, C. Eckerskorn, F. Lottspeich, K. Dietmeier, and N. Pfanner. 1996. Tom7 modulates the dynamics of the mitochondrial outer membrane translocase and plays a pathway-related role in protein import. EMBO J . 15:2125-2137. Huckaba, T.M., A.C. Gay, L.F. Pantalena, H.C. Yang, and L.A. Pon. 2004. Live cell imaging of the assembly, disassembly, and actin cable-dependent movement of endosomes and actin patches in the budding yeast, Saccharomyces cerevisiae. J Cell Biol . 167:519-530. Huckaba, T.M., T. Lipkin, and L.A. Pon. 2006. Roles of type II myosin and a tropomyosin isoform in retrograde actin flow in budding yeast. J Cell Biol . 175:957-969. Hwa Lim, H., F.M. Yeong, and U. Surana. 2003. Inactivation of mitotic kinase triggers translocation of MEN components to mother-daughter neck in yeast. Mol Biol Cell . 14:4734-4743. Itoh, T., E.A. Toh, and Y. Matsui. 2004. Mmr1p is a mitochondrial factor for Myo2p-dependent inheritance of mitochondria in the budding yeast. EMBO J . 23:2520-2530. Itoh, T., A. Watabe, E.A. Toh, and Y. Matsui. 2002. Complex formation with Ypt11p, a rab-type small GTPase, is essential to facilitate the function of Myo2p, a class V myosin, in mitochondrial distribution in Saccharomyces cerevisiae. Mol Cell Biol . 22:7744-7757. Jimenez, J., V.J. Cid, R. Cenamor, M. Yuste, G. Molero, C. Nombela, and M. Sanchez. 1998. Morphogenesis beyond cytokinetic arrest in Saccharomyces cerevisiae. J Cell Biol . 143:1617- 1634. Kakuda, T.N. 2000. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther . 22:685-708. Kasashima, K., M. Sumitani, M. Satoh, and H. Endo. 2008. Human prohibitin 1 maintains the organization and stability of the mitochondrial nucleoids. Exp Cell Res . 314:988-996. Koc, A., and G.F. Merrill. 2007. Checkpoint deficient rad53-11 yeast cannot accumulate dNTPs in response to DNA damage. Biochem Biophys Res Commun . 353:527-530. Kornmann, B., E. Currie, S.R. Collins, M. Schuldiner, J. Nunnari, J.S. Weissman, and P. Walter. 2009. An ER-Mitochondria Tethering Complex Revealed by a Synthetic Biology Screen. Science . 325:477- 481. Krapp, A., M.P. Gulli, and V. Simanis. 2004. SIN and the art of splitting the fission yeast cell. Curr Biol . 14:R722-730. Larosche, I., P. Letteron, B. Fromenty, N. Vadrot, A. Abbey-Toby, G. Feldmann, D. Pessayre, and A. Mansouri. 2007. Tamoxifen inhibits topoisomerases, depletes mitochondrial DNA, and triggers steatosis in mouse liver. J Pharmacol Exp Ther . 321:526-535. Lazzarino, D.A., I. Boldogh, M.G. Smith, J. Rosand, and L.A. Pon. 1994. Yeast mitochondria contain ATP- sensitive, reversible actin-binding activity. Mol Biol Cell . 5:807-818.

94

Lebedeva, M.A., and G.S. Shadel. 2007. Cell cycle- and ribonucleotide reductase-driven changes in mtDNA copy number influence mtDNA Inheritance without compromising mitochondrial gene expression. Cell Cycle . 6:2048-2057. Lee, H.C., P.H. Yin, J.C. Lin, C.C. Wu, C.Y. Chen, C.W. Wu, C.W. Chi, T.N. Tam, and Y.H. Wei. 2005. Mitochondrial genome instability and mtDNA depletion in human cancers. Ann N Y Acad Sci . 1042:109-122. Lee, S.J., M.F. Schwartz, J.K. Duong, and D.F. Stern. 2003. Rad53 phosphorylation site clusters are important for Rad53 regulation and signaling. Mol Cell Biol . 23:6300-6314. Lee, Y.S., W.D. Kennedy, and Y.W. Yin. 2009. Structural insight into processive human mitochondrial DNA synthesis and disease-related polymerase mutations. Cell . 139:312-324. Lippincott, J., and R. Li. 1998. Dual function of Cyk2, a cdc15/PSTPIP family protein, in regulating actomyosin ring dynamics and septin distribution. J Cell Biol . 143:1947-1960. Lippincott, J., K.B. Shannon, W. Shou, R.J. Deshaies, and R. Li. 2001. The Tem1 small GTPase controls actomyosin and septin dynamics during cytokinesis. J Cell Sci . 114:1379-1386. Liu, Z., and R.A. Butow. 2006. Mitochondrial retrograde signaling. Annu Rev Genet . 40:159-185. Longtine, M.S., A. McKenzie, 3rd, D.J. Demarini, N.G. Shah, A. Wach, A. Brachat, P. Philippsen, and J.R. Pringle. 1998. Additional modules for versatile and economical PCR-based gene deletion and modification in Saccharomyces cerevisiae. Yeast . 14:953-961. Luca, F.C., M. Mody, C. Kurischko, D.M. Roof, T.H. Giddings, and M. Winey. 2001. Saccharomyces cerevisiae Mob1p is required for cytokinesis and mitotic exit. Mol Cell Biol . 21:6972-6983. Luca, F.C., and M. Winey. 1998. MOB1, an essential yeast gene required for completion of mitosis and maintenance of ploidy. Mol Biol Cell . 9:29-46. Makovets, S., and E.H. Blackburn. 2009. DNA damage signalling prevents deleterious telomere addition at DNA breaks. Nat Cell Biol . 11:1383-1386. Mandal, S., P. Guptan, E. Owusu-Ansah, and U. Banerjee. 2005. Mitochondrial regulation of cell cycle progression during development as revealed by the tenured mutation in Drosophila. Dev Cell . 9:843-854. Matsushima, Y., R. Garesse, and L.S. Kaguni. 2004. Drosophila mitochondrial transcription factor B2 regulates mitochondrial DNA copy number and transcription in schneider cells. J Biol Chem . 279:26900-26905. McCammon, M.T., C.B. Epstein, B. Przybyla-Zawislak, L. McAlister-Henn, and R.A. Butow. 2003. Global transcription analysis of Krebs tricarboxylic acid cycle mutants reveals an alternating pattern of gene expression and effects on hypoxic and oxidative genes. Mol Biol Cell . 14:958-972. McKane, M., K.K. Wen, I.R. Boldogh, S. Ramcharan, L.A. Pon, and P.A. Rubenstein. 2005. A mammalian actin substitution in yeast actin (H372R) causes a suppressible mitochondria/vacuole phenotype. J Biol Chem . 280:36494-36501. Meeusen, S., and J. Nunnari. 2003. Evidence for a two membrane-spanning autonomous mitochondrial DNA replisome. J Cell Biol. 163:503-510. Meisinger, C., M. Rissler, A. Chacinska, L.K. Szklarz, D. Milenkovic, V. Kozjak, B. Schonfisch, C. Lohaus, H.E. Meyer, M.P. Yaffe, B. Guiard, N. Wiedemann, and N. Pfanner. 2004. The mitochondrial morphology protein Mdm10 functions in assembly of the preprotein translocase of the outer membrane. Dev Cell . 7:61-71. Meisinger, C., N. Wiedemann, M. Rissler, A. Strub, D. Milenkovic, B. Schonfisch, H. Muller, V. Kozjak, and N. Pfanner. 2006. Mitochondrial protein sorting: differentiation of beta-barrel assembly by Tom7-mediated segregation of Mdm10. J Biol Chem . 281:22819-22826. Meitinger, F., B. Petrova, I.M. Lombardi, D.T. Bertazzi, B. Hub, H. Zentgraf, and G. Pereira. 2010. Targeted localization of Inn1, Cyk3 and Chs2 by the mitotic-exit network regulates cytokinesis in budding yeast. J Cell Sci . 123:1851-1861.

95

Merkwirth, C., S. Dargazanli, T. Tatsuta, S. Geimer, B. Lower, F.T. Wunderlich, J.C. von Kleist-Retzow, A. Waisman, B. Westermann, and T. Langer. 2008. Prohibitins control cell proliferation and apoptosis by regulating OPA1-dependent cristae morphogenesis in mitochondria. Genes Dev . 22:476-488. Merkwirth, C., and T. Langer. 2009. Prohibitin function within mitochondria: essential roles for cell proliferation and cristae morphogenesis. Biochim Biophys Acta . 1793:27-32. Millard, P.J., B.L. Roth, H.P. Thi, S.T. Yue, and R.P. Haugland. 1997. Development of the FUN-1 family of fluorescent probes for vacuole labeling and viability testing of yeasts. Appl Environ Microbiol . 63:2897-2905. Mineri, R., N. Pavelka, E. Fernandez-Vizarra, P. Ricciardi-Castagnoli, M. Zeviani, and V. Tiranti. 2009. How do human cells react to the absence of mitochondrial DNA? PLoS One . 4:e5713. Moseley, J.B., and B.L. Goode. 2006. The yeast actin cytoskeleton: from cellular function to biochemical mechanism. Microbiol Mol Biol Rev . 70:605-645. Nishihama, R., J.H. Schreiter, M. Onishi, E.A. Vallen, J. Hanna, K. Moravcevic, M.F. Lippincott, H. Han, M.A. Lemmon, J.R. Pringle, and E. Bi. 2009. Role of Inn1 and its interactions with Hof1 and Cyk3 in promoting cleavage furrow and septum formation in S. cerevisiae. J Cell Biol . 185:995-1012. Nurse, P., Y. Masui, and L. Hartwell. 1998. Understanding the cell cycle. Nat Med . 4:1103-1106. O'Neill, B.M., S.J. Szyjka, E.T. Lis, A.O. Bailey, J.R. Yates, 3rd, O.M. Aparicio, and F.E. Romesberg. 2007. Pph3-Psy2 is a phosphatase complex required for Rad53 dephosphorylation and replication fork restart during recovery from DNA damage. Proc Natl Acad Sci U S A . 104:9290-9295. Osman, C., M. Haag, C. Potting, J. Rodenfels, P.V. Dip, F.T. Wieland, B. Brugger, B. Westermann, and T. Langer. 2009a. The genetic interactome of prohibitins: coordinated control of cardiolipin and phosphatidylethanolamine by conserved regulators in mitochondria. J Cell Biol . 184:583-596. Osman, C., C. Merkwirth, and T. Langer. 2009b. Prohibitins and the functional compartmentalization of mitochondrial membranes. J Cell Sci . 122:3823-3830. Owusu-Ansah, E., A. Yavari, S. Mandal, and U. Banerjee. 2008. Distinct mitochondrial retrograde signals control the G1-S cell cycle checkpoint. Nat Genet . 40:356-361. Park, C.B., and N.G. Larsson. 2011. Mitochondrial DNA mutations in disease and aging. J Cell Biol . 193:809-818. Paulovich, A.G., and L.H. Hartwell. 1995. A checkpoint regulates the rate of progression through S phase in S. cerevisiae in response to DNA damage. Cell . 82:841-847. Pellicioli, A., S.E. Lee, C. Lucca, M. Foiani, and J.E. Haber. 2001. Regulation of Saccharomyces Rad53 checkpoint kinase during adaptation from DNA damage-induced G2/M arrest. Mol Cell . 7:293- 300. Reichard, P. 1988. Interactions between deoxyribonucleotide and DNA synthesis. Annu Rev Biochem . 57:349-374. Schultz, R.A., S.J. Swoap, L.D. McDaniel, B. Zhang, E.C. Koon, D.J. Garry, K. Li, and R.S. Williams. 1998. Differential expression of mitochondrial DNA replication factors in mammalian tissues. J Biol Chem . 273:3447-3451. Schwartz, M.F., S.J. Lee, J.K. Duong, S. Eminaga, and D.F. Stern. 2003. FHA domain-mediated DNA checkpoint regulation of Rad53. Cell Cycle. 2:384-396. Senning, E.N., and A.H. Marcus. 2010. Actin polymerization driven mitochondrial transport in mating S. cerevisiae. Proc Natl Acad Sci U S A . 107:721-725. Seshan, A., and A. Amon. 2004. Linked for life: temporal and spatial coordination of late mitotic events. Curr Opin Cell Biol . 16:41-48. Sherman, F. 2002. Getting started with yeast. Methods Enzymol . 350:3-41.

96

Shou, W., J.H. Seol, A. Shevchenko, C. Baskerville, D. Moazed, Z.W. Chen, J. Jang, H. Charbonneau, and R.J. Deshaies. 1999. Exit from mitosis is triggered by Tem1-dependent release of the protein phosphatase Cdc14 from nucleolar RENT complex. Cell . 97:233-244. Sidorova, J.M., and L.L. Breeden. 1997. Rad53-dependent phosphorylation of Swi6 and down-regulation of CLN1 and CLN2 transcription occur in response to DNA damage in Saccharomyces cerevisiae. Genes Dev . 11:3032-3045. Simon, V.R., S.L. Karmon, and L.A. Pon. 1997. Mitochondrial inheritance: cell cycle and actin cable dependence of polarized mitochondrial movements in Saccharomyces cerevisiae. Cell Motil Cytoskeleton . 37:199-210. Singh, K.K., V. Ayyasamy, K.M. Owens, M.S. Koul, and M. Vujcic. 2009. Mutations in mitochondrial DNA polymerase-gamma promote breast tumorigenesis. J Hum Genet . 54:516-524. Sogo, L.F., and M.P. Yaffe. 1994. Regulation of mitochondrial morphology and inheritance by Mdm10p, a protein of the mitochondrial outer membrane. J Cell Biol . 126:1361-1373. Song, S., T.Z. Grenfell, S. Garfield, R.L. Erikson, and K.S. Lee. 2000. Essential function of the polo box of Cdc5 in subcellular localization and induction of cytokinetic structures. Mol Cell Biol . 20:286-298. Song, S., and K.S. Lee. 2001. A novel function of Saccharomyces cerevisiae CDC5 in cytokinesis. J Cell Biol . 152:451-469. Stegmeier, F., and A. Amon. 2004. Closing mitosis: the functions of the Cdc14 phosphatase and its regulation. Annu Rev Genet . 38:203-232. Taylor, S.D., H. Zhang, J.S. Eaton, M.S. Rodeheffer, M.A. Lebedeva, W. O'Rourke T, W. Siede, and G.S. Shadel. 2005. The conserved Mec1/Rad53 nuclear checkpoint pathway regulates mitochondrial DNA copy number in Saccharomyces cerevisiae. Mol Biol Cell . 16:3010-3018. Trifunovic, A., A. Wredenberg, M. Falkenberg, J.N. Spelbrink, A.T. Rovio, C.E. Bruder, Y.M. Bohlooly, S. Gidlof, A. Oldfors, R. Wibom, J. Tornell, H.T. Jacobs, and N.G. Larsson. 2004. Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature . 429:417-423. Tyynismaa, H., H. Sembongi, M. Bokori-Brown, C. Granycome, N. Ashley, J. Poulton, A. Jalanko, J.N. Spelbrink, I.J. Holt, and A. Suomalainen. 2004. Twinkle helicase is essential for mtDNA maintenance and regulates mtDNA copy number. Hum Mol Genet . 13:3219-3227. Tzagoloff, A., G. Macino, and W. Sebald. 1979. Mitochondrial genes and translation products. Annu Rev Biochem . 48:419-441. Van Aken, O., J. Whelan, and F. Van Breusegem. 2010. Prohibitins: mitochondrial partners in development and stress response. Trends Plant Sci . 15:275-282. Veses, V., A. Richards, and N.A. Gow. 2009. Vacuole inheritance regulates cell size and branching frequency of Candida albicans hyphae. Mol Microbiol . 71:505-519. Viikov, K., P. Valjamae, and J. Sedman. 2011. Yeast mitochondrial DNA polymerase is a highly processive single-subunit enzyme. Mitochondrion . 11:119-126. Wallace, D.C. 2010. Mitochondrial DNA mutations in disease and aging. Environ Mol Mutagen . 51:440- 450. Wang, Y., and D.F. Bogenhagen. 2006. Human mitochondrial DNA nucleoids are linked to protein folding machinery and metabolic enzymes at the mitochondrial inner membrane. J Biol Chem . 281:25791-25802. Weinert, T. 1998. DNA damage checkpoints update: getting molecular. Curr Opin Genet Dev . 8:185-193. Wiedemann, N., V. Kozjak, A. Chacinska, B. Schonfisch, S. Rospert, M.T. Ryan, N. Pfanner, and C. Meisinger. 2003. Machinery for protein sorting and assembly in the mitochondrial outer membrane. Nature . 424:565-571. Williamson, D.H., and D.J. Fennell. 1979. Visualization of yeast mitochondrial DNA with the fluorescent stain "DAPI". Methods Enzymol . 56:728-733.

97

Xu, S., H.K. Huang, P. Kaiser, M. Latterich, and T. Hunter. 2000. Phosphorylation and spindle pole body localization of the Cdc15p mitotic regulatory protein kinase in budding yeast. Curr Biol . 10:329- 332. Yang, H.C., and L.A. Pon. 2002. Actin cable dynamics in budding yeast. Proc Natl Acad Sci U S A . 99:751- 756. Yoshida, S., and A. Toh-e. 2001. Regulation of the localization of Dbf2 and mob1 during cell division of saccharomyces cerevisiae. Genes Genet Syst . 76:141-147. Youngman, M.J., A.E. Hobbs, S.M. Burgess, M. Srinivasan, and R.E. Jensen. 2004. Mmm2p, a mitochondrial outer membrane protein required for yeast mitochondrial shape and maintenance of mtDNA nucleoids. J Cell Biol . 164:677-688. Yu, M., Y. Zhou, Y. Shi, L. Ning, Y. Yang, X. Wei, N. Zhang, X. Hao, and R. Niu. 2007. Reduced mitochondrial DNA copy number is correlated with tumor progression and prognosis in Chinese breast cancer patients. IUBMB Life . 59:450-457. Zelenaya-Troitskaya, O., S.M. Newman, K. Okamoto, P.S. Perlman, and R.A. Butow. 1998. Functions of the high mobility group protein, Abf2p, in mitochondrial DNA segregation, recombination and copy number in Saccharomyces cerevisiae. Genetics . 148:1763-1776. Zhao, X., E.G. Muller, and R. Rothstein. 1998. A suppressor of two essential checkpoint genes identifies a novel protein that negatively affects dNTP pools. Mol Cell . 2:329-340. Zhao, X., and R. Rothstein. 2002. The Dun1 checkpoint kinase phosphorylates and regulates the ribonucleotide reductase inhibitor Sml1. Proc Natl Acad Sci U S A . 99:3746-3751.