11
84 Research Article Introduction Equal distribution of duplicated chromosomes during cell division is essential for maintaining genome integrity. During mitosis, accurate chromosome segregation is dependent on productive interactions between microtubules (MTs) and kinetochores (KTs), large proteinaceous structures that assemble onto centromeric DNA. Most errors in chromosome attachment activate the spindle assembly checkpoint (SAC), a surveillance mechanism that delays the onset of anaphase until chromosomes have attached to the mitotic spindle in a bipolar fashion. A major target of the SAC is Cdc20, a substrate-binding subunit of the APC/C (anaphase promoting complex/cyclosome). The APC/C is a large E3 ubiquitin ligase that initiates the ubiquitylation and subsequent degradation of key mitotic proteins, particularly cyclin B and securin (Musacchio and Salmon, 2007). Several critical components of this checkpoint were originally identified in budding yeast and include Mad1, Mad2, Mad3 (BubR1 in animals), Bub1, Bub3 and Mps1 (Musacchio and Salmon, 2007). These checkpoint proteins localize dynamically to unattached KTs. Elegant studies from several groups have collectively resulted in a model, according to which the KT provides an interface for the catalytic conversion of inactive, open-Mad2 to a closed-Mad2 capable of binding to Cdc20 and triggering the recruitment of BubR1-Bub3 into an APC/C inhibitory complex (Kulukian et al., 2009; Musacchio and Salmon, 2007; Yu, 2002). Although the exact composition of the resulting mitotic checkpoint complex (MCC) remains controversial, this complex is thought to shuttle off the KT as a diffusible ‘stop anaphase’ signal. This model is supported by the observation that the association between BubR1 and Cdc20 is dependent on Mad2 (Hardwick et al., 2000; Hwang et al., 1998; Nilsson et al., 2008), and it explains why both Mad2 and BubR1 are required for APC/C Cdc20 inhibition in vivo, although either protein is sufficient in vitro (Fang, 2002). SAC signaling appears to be more complex in animals than in yeast. Notably, BubR1 harbors a C-terminal Bub1-like kinase domain, and a second C-terminal Cdc20 binding region, both of which are lacking in the putative yeast ortholog Mad3 (Davenport et al., 2006; Harris et al., 2005; Tang et al., 2001). In addition to direct inhibition of APC/C Cdc20 , BubR1 contributes to the control of mitotic timing and the accurate chromosome capture by spindle microtubules (Ditchfield et al., 2003; Lampson and Kapoor, 2005). BubR1 is highly phosphorylated in mitosis, and we and others have recently shown that phosphorylation by Plk1 is important for maintaining proper kinetochore-microtubule (KT-MT) attachments and for timely mitotic progression (Elowe et al., 2007; Matsumura et al., 2007). More recently, additional (non-Plk1) phosphorylation sites have been identified, mutation of which also delayed mitotic exit (Huang et al., 2008). However, the identity of the kinase(s) responsible for phosphorylation of these sites remains unclear, although Mps1 might be involved. Insight into how BubR1 mediates APC/C Cdc20 inhibition came initially from studies in yeast, where it was demonstrated that two KEN boxes in Saccharomyces cerevisiae Mad3p are required for the SAC. These KEN motifs, which often mediate substrate recognition, are thought to inhibit the APC/C Cdc20 by competing with bona fide Cdc20 substrates (Burton and Solomon, 2007; King et al., 2007). The conservation of Mad3/BubR1 KEN motifs across evolution suggests that a similar mechanism may function in higher eukaryotes. Indeed, recent evidence suggested that N-terminal fragments of mouse BubR1 carrying both KEN boxes are sufficient Uncoupling of the spindle-checkpoint and chromosome-congression functions of BubR1 Sabine Elowe 1, *, Kalyan Dulla 1 , Andreas Uldschmid 1,‡ , Xiuling Li 2 , Zhen Dou 1 and Erich A. Nigg 1,3 1 Department of Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany 2 Group 1803, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, China 3 Biozentrum, University of Basel, 4056 Basel, Switzerland *Author for correspondence ([email protected]) Present address: Viramed Biotech AG, 82152 Planegg, Germany Accepted 21 October 2009 Journal of Cell Science 123, 84-94 Published by The Company of Biologists 2010 doi:10.1242/jcs.056507 Summary The BubR1 checkpoint protein performs multiple functions in mitosis. We have carried out a functional analysis of conserved motifs of human BubR1 (also known as BUB1B) and demonstrate that spindle assembly checkpoint (SAC) and chromosome attachment functions can be uncoupled from each other. Mutation of five proline-directed serine phosphorylation sites, identified in vivo by mass spectrometry, essentially abolishes attachment of chromosomes to the spindle but has no effect on SAC functionality. By contrast, mutation of the two conserved KEN boxes required for SAC function does not impact chromosome congression. Interestingly, the contribution of the two KEN-box motifs is not equal. Cdc20 associates with the N-terminal but not C-terminal KEN box, and mutation of the N-terminal KEN motif results in more severe acceleration of mitotic timing. Moreover, the two KEN motifs are not sufficient for maximal binding of Cdc20 and APC/C, which also requires sequences in the BubR1 C-terminus. Finally, mutation of the GLEBS motif causes loss of Bub3 interaction and mislocalization of BubR1 from the kinetochore; concomitantly, BubR1 phosphorylation as well as SAC activity and chromosome congression are impaired, indicating that the GLEBS motif is strictly required for both major functions of human BubR1. Key words: BubR1, Chromosome congression, Kinetochore, Mitosis, Spindle checkpoint Journal of Cell Science Journal of Cell Science

Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

84 Research Article

IntroductionEqual distribution of duplicated chromosomes during cell divisionis essential for maintaining genome integrity. During mitosis,accurate chromosome segregation is dependent on productiveinteractions between microtubules (MTs) and kinetochores (KTs),large proteinaceous structures that assemble onto centromericDNA. Most errors in chromosome attachment activate the spindleassembly checkpoint (SAC), a surveillance mechanism that delaysthe onset of anaphase until chromosomes have attached to themitotic spindle in a bipolar fashion. A major target of the SAC isCdc20, a substrate-binding subunit of the APC/C (anaphasepromoting complex/cyclosome). The APC/C is a large E3ubiquitin ligase that initiates the ubiquitylation and subsequentdegradation of key mitotic proteins, particularly cyclin B andsecurin (Musacchio and Salmon, 2007). Several criticalcomponents of this checkpoint were originally identified inbudding yeast and include Mad1, Mad2, Mad3 (BubR1 inanimals), Bub1, Bub3 and Mps1 (Musacchio and Salmon, 2007).These checkpoint proteins localize dynamically to unattached KTs.Elegant studies from several groups have collectively resulted ina model, according to which the KT provides an interface for thecatalytic conversion of inactive, open-Mad2 to a closed-Mad2capable of binding to Cdc20 and triggering the recruitment ofBubR1-Bub3 into an APC/C inhibitory complex (Kulukian et al.,2009; Musacchio and Salmon, 2007; Yu, 2002). Although the exactcomposition of the resulting mitotic checkpoint complex (MCC)remains controversial, this complex is thought to shuttle off theKT as a diffusible ‘stop anaphase’ signal. This model is supportedby the observation that the association between BubR1 andCdc20 is dependent on Mad2 (Hardwick et al., 2000; Hwang et

al., 1998; Nilsson et al., 2008), and it explains why both Mad2and BubR1 are required for APC/CCdc20 inhibition in vivo,although either protein is sufficient in vitro (Fang, 2002).

SAC signaling appears to be more complex in animals than inyeast. Notably, BubR1 harbors a C-terminal Bub1-like kinasedomain, and a second C-terminal Cdc20 binding region, both ofwhich are lacking in the putative yeast ortholog Mad3 (Davenportet al., 2006; Harris et al., 2005; Tang et al., 2001). In addition todirect inhibition of APC/CCdc20, BubR1 contributes to the controlof mitotic timing and the accurate chromosome capture by spindlemicrotubules (Ditchfield et al., 2003; Lampson and Kapoor, 2005).BubR1 is highly phosphorylated in mitosis, and we and others haverecently shown that phosphorylation by Plk1 is important formaintaining proper kinetochore-microtubule (KT-MT) attachmentsand for timely mitotic progression (Elowe et al., 2007; Matsumuraet al., 2007). More recently, additional (non-Plk1) phosphorylationsites have been identified, mutation of which also delayed mitoticexit (Huang et al., 2008). However, the identity of the kinase(s)responsible for phosphorylation of these sites remains unclear,although Mps1 might be involved.

Insight into how BubR1 mediates APC/CCdc20 inhibition cameinitially from studies in yeast, where it was demonstrated that twoKEN boxes in Saccharomyces cerevisiae Mad3p are required forthe SAC. These KEN motifs, which often mediate substraterecognition, are thought to inhibit the APC/CCdc20 by competingwith bona fide Cdc20 substrates (Burton and Solomon, 2007; Kinget al., 2007). The conservation of Mad3/BubR1 KEN motifs acrossevolution suggests that a similar mechanism may function in highereukaryotes. Indeed, recent evidence suggested that N-terminalfragments of mouse BubR1 carrying both KEN boxes are sufficient

Uncoupling of the spindle-checkpoint andchromosome-congression functions of BubR1Sabine Elowe1,*, Kalyan Dulla1, Andreas Uldschmid1,‡, Xiuling Li2, Zhen Dou1 and Erich A. Nigg1,3

1Department of Cell Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany2Group 1803, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, China3Biozentrum, University of Basel, 4056 Basel, Switzerland*Author for correspondence ([email protected])‡Present address: Viramed Biotech AG, 82152 Planegg, Germany

Accepted 21 October 2009Journal of Cell Science 123, 84-94 Published by The Company of Biologists 2010doi:10.1242/jcs.056507

SummaryThe BubR1 checkpoint protein performs multiple functions in mitosis. We have carried out a functional analysis of conserved motifsof human BubR1 (also known as BUB1B) and demonstrate that spindle assembly checkpoint (SAC) and chromosome attachmentfunctions can be uncoupled from each other. Mutation of five proline-directed serine phosphorylation sites, identified in vivo by massspectrometry, essentially abolishes attachment of chromosomes to the spindle but has no effect on SAC functionality. By contrast,mutation of the two conserved KEN boxes required for SAC function does not impact chromosome congression. Interestingly, thecontribution of the two KEN-box motifs is not equal. Cdc20 associates with the N-terminal but not C-terminal KEN box, and mutationof the N-terminal KEN motif results in more severe acceleration of mitotic timing. Moreover, the two KEN motifs are not sufficientfor maximal binding of Cdc20 and APC/C, which also requires sequences in the BubR1 C-terminus. Finally, mutation of the GLEBSmotif causes loss of Bub3 interaction and mislocalization of BubR1 from the kinetochore; concomitantly, BubR1 phosphorylation aswell as SAC activity and chromosome congression are impaired, indicating that the GLEBS motif is strictly required for both majorfunctions of human BubR1.

Key words: BubR1, Chromosome congression, Kinetochore, Mitosis, Spindle checkpoint

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 2: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

85Uncoupling of BubR1 mitotic functions

for cell survival, whereas mutation of either KEN box led to lossof Cdc20 binding and cell viability (Malureanu et al., 2009).

Here, we examine the contribution of conserved motifs to themitotic functions of full-length human BubR1. We identify severalconserved phosphorylation sites in BubR1 and demonstrate thatCdk1 (CDK1) rather than Mps1 (also known as TTK)phosphorylation contributes significantly to KT-MT attachments,but not to MCC formation or other SAC functions. Conversely, cellsexpressing BubR1 KEN-box mutants exit mitosis prematurely andare unable to arrest in response to nocodazole treatment. Such cellsare nevertheless capable of efficiently aligning their chromosomecomplement, provided that they are maintained in mitosis by MG132treatment. Furthermore, we demonstrate that efficient Cdc20 andAPC/C binding requires full-length BubR1, as N-terminal fragmentsof BubR1 bound poorly to both Cdc20 and the APC/C. These datasuggest that the chromosome congression and checkpoint functionsof BubR1 can be uncoupled from each other. Interestingly, a BubR1GLEBS domain mutant, which is unable to bind Bub3 and showsreduced accumulation at the KT, no longer becomes detectablyphosphorylated. Strikingly, this mutant exhibits both congressionand checkpoint defects, highlighting the importance of the BubR1GLEBS domain for its mitotic functions.

ResultsIdentification and quantification of BubR1phosphorylation sites in mitosisIt was recently shown that phosphorylation of BubR1 by Plk1 isrequired for stable KT-MT attachments (Elowe et al., 2007;Matsumura et al., 2007). Nevertheless, Plk1 phosphorylation cannotalone account for the congression defects observed in BubR1-depleted cells (Lampson and Kapoor, 2005). Indeed, in addition toPlk1, Cdk1 is able to efficiently phosphorylate BubR1 in vitro(Elowe et al., 2007). We therefore sought to identify novelphosphorylation sites on BubR1 that are specifically upregulatedin mitosis. Using mass spectrometry, we identified mitotic-specificphosphorylation at the serine-proline (SP) motifs around S543,S574, S670, S720 and S1043 (Fig. 1A and supplementary materialFig. S1A), as well as 12 other residues. Using the stable isotopelabeling with amino acids in cell culture (SILAC) approach, wewere able to quantify the extent of mitotic-specific phosphorylationat four of these sites (supplementary material Fig. S1B), and showthat phosphorylation is upregulated during nocodazole arrest (Fig.1A). S574 was identified in a non-tryptic peptide; as it containedneither arginine nor lysine it could not be directly quantified by theSILAC procedure used here. Independently, Huang et al. recentlyreported the identification of S543, S670, and S1043 as mitotic-specific phosphorylation sites on BubR1 (Huang et al., 2008), butS720 and S574 are novel sites.

To test whether the phosphorylation sites identified herecontribute to the characteristic electrophoretic upshift of mitoticBubR1, MYC-tagged constructs for BubR1-WT, BubR1-620A(Polo-box domain binding mutant), BubR1-5A, and thephosphomimetic 5D (where all five phosphorylation sites weremutated to aspartate) were expressed in HeLa cells. The S620Amutant was included in this analysis, as we have previously shownthat Plk1 phosphorylation regulates the characteristic mitotic upshiftin BubR1 (Elowe et al., 2007). Cells were then harvested after a16-hour release from thymidine arrest into nocodazole, beforeproteins were analyzed by western blotting. BubR1-WT exhibitedthe characteristic double band pattern, whereas BubR1-620A, aspredicted, migrated as a single faster migrating species (Fig. 1B).

Both BubR1-5A and BubR1-5D also exhibited a double bandpattern, suggesting that mutation of these five residues is notsufficient to eliminate the mitotic BubR1 upshift (Fig. 1B).

Interestingly, the five sites described here consist of a serinefollowed by a proline residue, suggesting that a proline-directedkinase such as Cdk1 may phosphorylate these sites (Fig. 1C),although phosphorylation at several of these has been suggested tobe Mps1-dependent (Huang et al., 2008). To compare BubR1phosphorylation by Cdk1 and Mps1, we performed in vitro kinaseassays using recombinant full-length BubR1 as a substrate (Fig.1D). Whereas Cdk1 was able to efficiently phosphorylate BubR1,there was no detectable phosphorylation of BubR1 by Mps1 kinaseat the same specific activity (Fig. 1D, left panels), although bothCdk1 and Mps1 efficiently phosphorylated Borealin under the sameconditions (Fig. 1D, right panels). To clarify whether the SP sitesidentified here are indeed Cdk1 target sites, we generated 12-mer

Fig. 1. Identification of BubR1 in vivo phosphorylation sites.(A)Phosphosites identified by mass spectrometry. The table shows the resultsfrom one representative experiment. The exact peptides identified are listedalong with the position of the phosphosite, MASCOT score, the M phase:Sphase (M/S) ratio of phosphorylated peptide as determined by SILAC, and theM/S protein ratio. (B)Western blot showing the electrophoretic mobility ofMYC-tagged BubR1-WT and the phosphosite mutants BubR1-620A, -5A, and-5D (upper panel). a-Tubulin was used as a loading control from total celllysate (TCL). (C)Sequence alignment of the five identified phosphorylationsites, indicating conservation of the SP motif in these sites in BubR1 (red box).(D)In vitro phosphorylation of recombinant MBP-BubR1 (left panels) orMBP-Borealin (right panels) in either kinase buffer alone, or with Cdk1 orMps1. BubR1 and Borealin were identified by Coomassie Brilliant Blue(CBB) staining (lower panels). (E)Immobilized peptides synthesized directlyon cellulose membranes were used as substrates for Cdk1 in in vitro kinaseassays. Peptide phosphorylation was visualized by autoradiography.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 3: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

86

peptides centered on each of the five serines and used these peptidesas in vitro substrates for Cdk1. Corresponding control peptides weresynthesized with the serine phospho-acceptor positions changed toalanine to determine signal specificity. The peptides that includedS543, S670 and S1043 became phosphorylated in this assay,whereas the alanine versions of the same peptides were either notdetectably phosphorylated or phosphorylated to a significantlyreduced extent (Fig. 1E). Collectively, these observations suggestthat Cdk1 may directly phosphorylate BubR1 in vivo, and that theloss of BubR1 phosphorylation upon Mps1 depletion may reflectan indirect mechanism.

BubR1-5A-mutant-expressing cells exhibit severecongression defectsInitially we sought to determine whether loss of BubR1phosphorylation at the SP sites identified here results in changes toBubR1 localization or in gross structural defects in the protein.Endogenous BubR1 was depleted using siRNA oligos that targetthe 3�-UTR region of BubR1 (supplementary material Fig. S2)(Elowe et al., 2007), and cells were simultaneously transfected withMYC-tagged BubR1-WT or phosphorylation site mutants.MYC–BubR1-WT localized as expected to the outer-KT, asdemonstrated by colocalization with Hec1 (supplementary materialFig. S3A), as did MYC–BubR1-620A, -5A and -5D. In additionBubR1-5A and -5D retained the ability to coimmunoprecipitateCdc20 and Bub3 (supplementary material Fig. S3B), confirmingthat MCC assembly is maintained. Finally, mutation of the five SPsites did not affect phosphorylation of BubR1 at the previouslyreported S676 site of Plk1 (supplementary material Fig. S3C). Theseresults demonstrate that mutation of the five phosphorylation siteson BubR1 did not result in gross conformational or structural defectsin the BubR1 protein that would preclude KT localization and MCCformation.

To explore whether the phosphorylation of the five identified SPsites on BubR1 contributes to the congression or checkpointfunction of BubR1 (or both), we took several independentapproaches. Initially, we examined mitotic progression by stainingof fixed cells (Fig. 2A,B). After MG132 treatment, BubR1-620A,-5A, and -5D-expressing cells were all unable to align propermetaphases to the same extent as BubR1-WT-expressing cells(supplementary material Fig. S4A). This indicates that lack ofphosphorylation on one or several of the five sites identified in thisstudy causes congression defects, and that the BubR1-5Dphosphomimic mutant cannot rescue the BubR1 depletionphenotype. A BubR1-4A/D mutant (which retained the previouslydescribed S670) also resulted in aberrant metaphases after MG132treatment, indicating a significant contribution from the remainingfour serine residues to the chromosome alignment function ofBubR1 (supplementary material Fig. S4B).

Close examination of cells expressing the various mutantsrevealed differences with regard to their precise congression defects.As expected, cells depleted of endogenous BubR1 and rescued withthe empty plasmid failed to congress chromosomes efficiently, sothat KTs were spread along the length of the pole-to-pole axis (Fig.2B). By contrast, cells expressing BubR1-WT generally formed tightmetaphase plates, with KTs efficiently aligned at the spindleequator. Cells expressing BubR1-620A were largely able to aligntheir KTs, although resulting metaphases were considerably broader,whereas both BubR1-5A- and -5D-expressing cells resembledthose depleted of endogenous BubR1, with many KTs remainingunattached and spread along the entire pole-to-pole axis (Fig. 2B,

Journal of Cell Science 123 (1)

supplementary material Fig. S4C for MYC construct expression).Quantification of KT misalignment with the various BubR1 mutantsrevealed that the degree of misalignment in cells expressing eitherBubR1-5A or BubR1-5D was almost as severe as in cells entirelydepleted of BubR1 (25% in BubR1-5A and 22% in BubR1-5Dcompared to 30% of cells transfected with control vector; Fig. 2C).These observations suggest that these five phosphorylation sitesprobably make the most significant contribution to the congressionfunction of BubR1.

As a second, complementary approach, we examined the functionof BubR1 phosphorylation site mutants in real-time by time-lapsevideo microscopy performed on HeLa cells stably expressinghistone H2B-GFP. To track individual cells, we used BubR1constructs tagged with mCherry, as previously described (Elowe etal., 2007). Representative stills from each movie, taken at theindicated time points after the onset of chromosome condensation,are shown in Fig. 2D, and quantification of time in mitosis issummarized in Fig. 2E. Cells depleted of endogenous BubR1 andexpressing the mCherry tag alone exited mitosis very rapidly (90%of the cells within 60 minutes, supplementary material Movie 1),without metaphase alignment and often with lagging chromosomes,as expected. This phenotype was rescued by BubR1-WT expression(supplementary material Movie 2). As described previously, BubR1-620A-expressing cells exited mitosis more slowly, withapproximately 40% of the cells entering anaphase more than 140minutes after chromosome condensation, compared with about 25%of BubR1-WT expressing cells (supplementary material Movie 3).Cells expressing BubR1-5A or BubR1-5D displayed severecongression defects (supplementary material Movies 4 and 5,respectively); they were often unable to reach metaphase and manycells were unable to exit mitosis within the imaging period (16hours). As a result, only 25% of BubR1-5A- and 42% of BubR1-5D-expressing cells reached anaphase within 140 minutes.Interestingly, amongst the cells expressing BubR1-5A and BubR1-5D that were able to exit mitosis, we were unable to detect anylagging chromosomes in anaphase. This, together with theobservation that the many cells unable to align at metaphase didnot exit mitosis, suggests that the checkpoint function of BubR1 ismaintained in the BubR1-5A mutant.

To more rigorously test this conclusion, we assessed the abilityof cells expressing either MYC–BubR1-WT or BubR1 phosphositemutants to remain mitotically arrested upon microtubuledepolymerization. Non-rescued cells were unable to arrest in thepresence of nocodazole, whereas cells rescued with BubR1-WTarrested efficiently (Fig. 2F). Similarly, cells expressingMYC–BubR1-620A, -5A or -5D all arrested to the same extent asthose expressing BubR1-WT, indicating that the SAC function ofBubR1 is functional in these cells. Collectively, our observationsindicate that phosphorylation at the five sites described here iscritical for BubR1 function in KT-MT attachment and chromosomecongression, but is not required for mediating the SAC response ineither an unperturbed mitosis or after a nocodazole challenge.

Interplay between BubR1 S670 and S676 phosphorylationS670 and S676 are the best-conserved of the BubR1 phosphorylationsites identified to date. Indeed many residues in this region of theprotein are conserved in higher eukaryotes (Fig. 3A). To study S670phosphorylation, we generated an anti-pS670 antibody.Phosphospecificity of the antibody was demonstrated by loss ofreactivity after phosphatase treatment as seen byimmunofluorescence and western blotting (supplementary material

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 4: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

87Uncoupling of BubR1 mitotic functions

Fig. S5A,B). Furthermore, antibody reactivity in cells depleted ofendogenous BubR1 could be restored by MYC–BubR1-WT but notthe non-phosphorylatable MYC–BubR1-670A (supplementarymaterial Fig. S5C). Analysis of an asynchronous cell populationby immunofluorescence microscopy revealed that BubR1 S670 wasphosphorylated predominantly at unaligned KTs duringprometaphase and in cells nearing metaphase (supplementarymaterial Fig. S5D). Anti-pS670 was also observed to decorate thealigned KTs of metaphase plates (supplementary material Fig. S5D),which contrasts with the results obtained with the anti-pS676antibody (Elowe et al., 2007). This suggests that phosphorylationat S670 may be important for KT attachments throughout mitosis.

To test the effect of alterations in microtubule dynamics on S670phosphorylation, we treated mitotic cells with DMSO, nocodazole,or Taxol for 30 minutes before fixation. Cells were then probed for

S670 and S676 phosphorylation by immunofluorescence using thecorresponding phosphospecific antibodies. Focusing onprometaphase cells we observed only minor differences in S676phosphorylation between conditions tested (data not shown, seequantification in Fig. 3C), consistent with the lack of inter-KTtension in prometaphase. By contrast, S670 phosphorylation wassignificantly elevated in prometaphase cells treated with 300 nMnocodazole, as compared to the DMSO control, whereas no increasein phosphorylation was detected after treatment with 100 nM Taxol(Fig. 3B, and quantification in C), even though this dose of Taxolefficiently induces rephosphorylation of S676 when cells arealigned at metaphase (Elowe et al., 2007). In agreement with thesefindings, a marked increase in pS670 reactivity after nocodazolebut not Taxol or DMSO treatment was observed by western blotting(Fig. 3D). These results indicate that S670 phosphorylation is

Fig. 2. BubR1-5A mutants cause severecongression defects but retain SAC function.(A)Schematic representation of BubR1 rescueprotocol for testing mitotic progression andchromosome alignment. T, time in hours; thym,thymidine. (B)Representative images ofmetaphase cells expressing the various BubR1phosphomutants after 3 hours of MG132treatment. Cells were stained for a-tubulin(Cy2, shown in green), CREST humanautoimmune serum (Cy5, shown here in red)and MYC (Cy3; shown in red in Fig. S3C) tofacilitate identification of rescued cells.Representative KT-MT connections areindicated by white arrows, and enlarged imagesand are shown below. Scale bar: 10m.(C)Quantification of the degree of chromosomemisalignment in cells expressing BubR1mutants, shown in B. KT position was measuredas a function of the distance from pole to spindleequator. Values are mean ± s.e.m. of five to sixcells; >60 KTs per cell. (D)Representative stillsfrom time-lapse videomicroscopy experimentsillustrating mitotic progression of HeLa cellsstably expressing histone H2B-GFP depleted ofendogenous BubR1 and rescued with mCherryempty vector, mCherry-BubR1-WT,-620A, -5Aor -5D. Images were acquired at the indicatedtimepoints (hours:minutes) after the start ofchromosome condensation. (E)Bar graphindicating time elapsed between chromosomecondensation and anaphase onset of the cells inD. Elapsed time was split into three categories:0-60 minutes, 60-140 minutes, and >140minutes, and the percentage of cells expressingthe different MYC-BubR1 constructs wasplotted for each time category. Values are mean± s.e.m. of eight independent experiments(n100-220 cells). (F)Bar graph showingmitotic index (after 14 hours nocodazoletreatment) in cells depleted of endogenousBubR1, and expressing BubR1 phosphositemutants relative to BubR1-WT-expressingcells. Values are mean ± s.e.m. of threeindependent experiments (160-250 cells perexperiment). The mitotic index of non-treatedcell analyzed under the same conditions was27%.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 5: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

88

elevated at unattached rather than tensionless KTs. A similarconclusion was recently reached by Yen and co-workers, using anindependently generated antibody against pS670 (Huang et al.,2008).

As S676 phosphorylation is lost at KTs upon metaphasealignment and establishment of tension, we considered thepossibility that the phosphorylation status of S670 may regulateS676 phosphorylation by Plk1 at this stage. Indeed, when cells weremaintained at metaphase by MG132 treatment, S676phosphorylation was still detectable in BubR1-S670A or BubR1-S670D expressing cells, although, as expected, it was lost in BubR1-WT expressing cells (Fig. 3E). This indicates that the KT-MTattachments in cells expressing BubR1-S670A/D were unable togenerate sufficient tension at metaphase to turn off S676phosphorylation by Plk1, although we cannot directly excludemicroenvironment alterations that effect anti-pS676 reactivity inresponse to changes in S670 phosphorylation.

Investigation of the BubR1-Cdc20 interactionIn addition to phosphorylation sites, BubR1 and Mad3 proteinscontain several evolutionarily conserved motifs. These include twoKEN boxes and a GLEBS-like motif which is required for Bub3binding and Mad3 KT localization. In S. cerevisiae andSchizosaccharomyces pombe, the direct interaction betweenMad3p and Cdc20p is mediated through the N-terminal, but notC-terminal KEN box (Burton and Solomon, 2007; King et al.,2007; Sczaniecka et al., 2008), whereas a recent study hassuggested that both N- and C-terminal KEN motifs of murineBubR1 are required for association with Cdc20 (Malureanu et al.,2009). To resolve this discrepancy, we initially generatedimmobilized peptides encompassing either N-terminal (KEN26)or C-terminal (KEN304) KEN-box motifs and tested their ability

Journal of Cell Science 123 (1)

to directly bind recombinant GST-Cdc20 or GST alone. Whereasthe KEN26 motif of human BubR1 bound efficiently torecombinant Cdc20, a peptide encompassing the KEN304 boxshowed no binding when assayed under the same conditions (Fig.4A). Neither peptide was able to associate with GST alone,demonstrating the specificity of the interaction. This is in contrastto a recent report on mBubR1 (Malureanu et al., 2009), but in fullagreement with data from S. cerevisiae and S. pombe (King et al.,2007; Sczaniecka et al., 2008).

How the KEN motifs function in the context of the full-lengthBubR1 molecule is not clear, as BubR1 binding to Cdc20 involvesmultiple sites, and previous reports only focused on truncated N-terminal BubR1 (Davenport et al., 2006; Malureanu et al., 2009).We therefore asked whether various full-length human BubR1proteins carrying point mutations in these critical motifs are ableto form a complex with other MCC components and the APC/C inmitosis. Endogenous Cdc20, Bub3 and Cdc27 could readily be co-immunoprecipitated with MYC–BubR1-WT, as expected (Fig.4B). Similar results were observed after immunoprecipitation ofMYC-tagged BubR1-620A and BubR1-5A, indicating that MCCformation is intact in the BubR1 phosphorylation site mutants.Surprisingly, we found that full-length BubR1-KEN26AAA as wellas BubR1-KEN304AAA associated with appreciable amounts ofCdc20, Cdc27 and Bub3 (Fig. 4B), and even mutation of bothBubR1 KEN boxes together (BubR1-KEN) did not abrogate theinteraction with these proteins (Fig. 4C, supplementary material Fig.S6A). Therefore, in the context of full-length BubR1, mutation ofKEN26 alone or in conjunction with KEN304 is not sufficient toeliminate the interaction with Cdc20 and the APC/C, probablybecause of the presence of a second C-terminal Cdc20 bindingregion in BubR1 (Davenport et al., 2006; Tang et al., 2001). Forcomparison, BubR1-E413K, carrying a mutation in the Bub3

Fig. 3. Characterization of BubR1 phosphorylation atS670. (A)Sequence alignment of a central region inBubR1 showing conservation of S670 and S676 inmetazoans. Hs, Homo sapiens; Mm, Mus musculus; Gg,Gallus gallus; Xl, Xenopus laevis; Dr, Danio rerio; Dm,Drosophila melanogaster. (B)S670 phosphorylation iselevated in response to nocodazole but not Taxoltreatment. HeLa cells were released for 10 hours from athymidine block before being treated with DMSO, Taxolor nocodazole for 30 minutes. Cells were then fixed andstained with antibodies against pS670 (red), BubR1 (blue)and a-tubulin (green). DNA was visualized with DAPI.(C)Quantification of the levels of S670 and S676phosphorylation (relative to total BubR1) inprometaphase cells treated as in B. *P<0.03 versusDMSO-treated cells. (D)Anti-pS670 (top panel) and anti-pS676 (middle panel) were used in western blotting onimmunoprecipitates of endogenous BubR1 from cellstreated as in B. The same blot was stripped and reprobedwith anti-BubR1 antibodies to demonstrate equal input.(E)HeLa cells were simultaneously depleted ofendogenous BubR1 and rescued with MYC-taggedBubR1-WT, -670A or -670D, released from a thymidineblock for 10 hours into mitosis, and treated a further 3hours with MG132 before being fixed and stained withanti-pS676 antibodies (red), and anti-MYC (green). Scalebar: 10m.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 6: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

89Uncoupling of BubR1 mitotic functions

binding (GLEBS) region also co-immunoprecipitated Cdc20 andCdc27, but, as expected, not Bub3 (Fig. 4B).

To directly compare the efficiency of Cdc20 binding betweenfull-length and N-terminal BubR1, we generated an N-terminalfragment of human BubR1 (BubR1-N, residues 1-370) in its WTform or with one or both KEN motifs mutated, and tested the abilityof these fragments to bind Cdc20 and the APC/C. Full-length

BubR1-WT bound efficiently to Cdc20, Cdc27, and Bub3, asexpected, whereas BubR1-N-WT recruited Cdc20 at much lowerlevels (Fig. 4D, supplementary material Fig. S6B). This interactionwas completely eliminated in BubR1-N-KEN26AAA, and BubR1-N-KEN, whereas BubR1-N-KEN304AAA bound Cdc20 at levelscomparable to BubR1-N-WT. The full complement of Cdc20binding therefore requires the BubR1 C-terminus; the N-terminusof BubR1 can only recruit low levels of Cdc20 through KEN26(but not KEN304), in agreement with our peptide binding studiesand observations in yeast (King et al., 2007; Sczaniecka et al., 2008).Moreover, in the absence of sufficient Cdc20 binding, BubR1 isunable to efficiently associate with the APC/C. Indeed, upon siRNA-mediated depletion of Cdc20, BubR1 immunoprecipitates containedreduced levels of the APC/C subunits Cdc27, Apc7 and Apc4compared with control cells (Fig. 4E).

Characterization of BubR1 checkpoint defects in KEN boxand GLEBS mutantsBubR1 mutated in either the N-terminal or C-terminal KEN boxefficiently localized to the KT in cells depleted of endogenousBubR1, whereas BubR1-E413K was unable to accumulate at KTs,as predicted (Fig. 5A, for quantification see supplementary materialFig. S7). Recently, it was shown that BubR1 interacts with Blinkinvia N-terminal TPR motifs and independent of Bub3 (Kiyomitsuet al., 2007). The relevant region is conserved in Bub1 andmutations in this region abrogate Bub1 KT recruitment (Kiyomitsuet al., 2007). By contrast, we find that the Blinkin-binding regionof BubR1 (residues 1-203) was largely cytoplasmic, indicating thatBlinkin binding alone is not sufficient for KT localization of BubR1(supplementary material Fig. S7).

We next sought to determine whether SAC function wasperturbed in full-length BubR1 lacking functional KEN and KTrecruitment motifs. Having shown that full-length BubR1 mutatedat either KEN box was still able to associate with significant amountsof Cdc20, we asked whether cells expressing these mutants wouldbe able to mount a SAC response. However, expression of BubR1-KEN26AAA or -KEN304AAA in cells depleted of endogenousBubR1 was not sufficient to sustain a mitotic arrest in the presenceof nocodazole (Fig. 5B), in full agreement with observations in yeastand mouse. These data underscore the notion that Cdc20 bindingalone is not sufficient for APC/CCdc20 inhibition by BubR1.Interestingly, HeLa cells expressing BubR1-E413K were equallydefective in arresting in response to a nocodazole (Fig. 5B),suggesting that Bub3 binding and/or BubR1 localization to KTs isrequisite for SAC function.

To study the role of the KEN and GLEBS motifs in regulatingmitotic timing, we turned to time-lapse videomicroscopy of HeLacells stably expressing histone H2B-GFP. Expression of BubR1-KEN26AAA, -KEN304AAA, or -E413K failed to restore thetimely and accurate mitotic progression that occurred withexpression of BubR1-WT, but interesting differences in phenotypeswere observed. BubR1-KEN26AAA- and -E413K-expressing cellsexited mitosis with kinetics very similar to cells depleted ofendogenous BubR1 (with a median mitotic time of 24±1 and 30±4minutes, respectively, see Fig. 5D and supplementary materialMovies 6 and 7) and often exhibited misaligned chromosomes uponanaphase onset, indicative of a defective SAC (see arrowhead).By contrast, although cells expressing BubR1-KEN304AAA alsoexited mitosis faster than BubR1-WT-expressing cells (median45±2 minutes, see Fig. 5D and supplementary material Movie 8),in each of three independent experiments they were consistently

Fig. 4. Investigation of the interaction between BubR1 and Cdc20.(A)Immobilized peptides corresponding to KEN26 and KEN304 were directlysynthesized onto cellulose membranes, and were probed with recombinantGST-Cdc20 or GST protein. Peptide sequences are described in the Materialsand Methods. (B)MYC-tagged BubR1-WT, BubR1-620A, BubR1-5A,BubR1-KEN26AAA, BubR1-KEN304AAA and BubR1-E413K wereexpressed in HEK293T cells. After 36 hours, cells were treated withnocodazole for a further 16 hours and subsequently harvested. MYC-BubR1was then immunoprecipitated using anti-MYC antibodies, resolved by SDS-PAGE, and coimmunoprecipitation of Cdc20, Cdc27, and Bub3 was resolvedby western blotting. The same membrane was reprobed for total MYC. Equalinput was verified by blotting for a-tubulin from total cell lysates (TCL).(C)MYC-tagged BubR1-WT, -KEN26AAA, -KEN304AAA, or -KEN wereexpressed in HEK 293T cells, and co-immunoprecipiation of Cdc20, Cdc27,and Bub3 was resolved as in (B). (D)MYC-tagged full-length BubR1-WT andBubR1-N (residues 1-370), either WT or the KEN-box mutant forms, weretransfected into HEK293T cells, and coimmunoprecipitation of Cdc20, Cdc27and Bub3 was resolved as in B. (E)HeLa cells depleted of Cdc20 by siRNA ortreated with control siRNA were released from a thymidine block for 9 hoursand treated for a further 3 hours with MG132 to enrich for cells in mitosis.Cells were collected, and lysates from each condition were normalized forprotein content before immunoprecipitation of BubR1. Samples were thenresolved by SDS-PAGE and blotted for coimmunoprecipitation of Cdc20,Cdc27, Apc7, Apc4 and Bub3. Equal protein input is demonstrated byreprobing for BubR1 in the immunoprecipitate, and by blotting for Bub3 intotal cell lysate (TCL). Efficient Cdc20 depletion is shown by western blottingof TCL.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 7: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

90

slower than either BubR1-KEN26AAA- or BubR1-E413K-expressing cells, or cells depleted of BubR1 (P0.03 forKEN304AAA-expressing vs BubR1-depleted cells). Moreover, thechromosomes of these cells were often observed to align atmetaphase, although alignment was not maintained for the sameduration as in BubR1-WT-expressing cells (Fig. 5C). Similar resultswere obtained upon analysis of fixed cells by immunofluorescencemicroscopy. Cells expressing the control empty vector orexpressing BubR1-KEN26AAA, -KEN304AAA or -E413K exitedmitosis prematurely compared with BubR1-WT-expressing cells,as demonstrated by the larger proportion of transfected cells inanaphase (Fig. 5E). Together, these results demonstrate that, in thecontext of full-length human BubR1, KEN26, KEN304 and theGLEBS motif are all essential for SAC function and, to varyingextents, for timely mitotic progression.

Journal of Cell Science 123 (1)

Cells expressing BubR1 KEN26AAA or KEN304AAA canachieve normal metaphaseThe difference in mitotic progression time between cells expressingBubR1-KEN304AAA and other checkpoint mutants tested here (onaverage 15 minutes), prompted us to question whether this extratime contributed to the correction of chromosome attachment errorsthat occur frequently in early mitosis. We therefore explored theextent to which chromosomes were found lagging in anaphase cellsexpressing the various BubR1 SAC mutants in depletion-rescueexperiments. Less than 20% of cells depleted of endogenousBubR1 exhibited normal anaphases, with the vast majority showingmore than two lagging KTs, whereas the majority of cells expressingBubR1-WT exited mitosis accurately (Fig. 6A,B). Cells expressingBubR1-KEN26AAA or BubR1-E413K exhibited lagging KTs inanaphase at levels significantly higher than those expressing BubR1-

Fig. 5. Characterization of SAC defectscaused by BubR1 KEN-box and GLEBSdomain mutants. (A)HeLa cells were depletedof endogenous BubR1 and rescued with MYC-tagged BubR1-WT, MYC–BubR1-26AAA,MYC–BubR1-304AAA or MYC–BubR1-E413K. After release from a thymidine blockinto mitosis for 10 hours, cells were fixed andstained with antibodies against MYC (red), andBub1 (green). DNA was visualized with DAPI.Scale bar: 10m. (B)Bar graph showing themitotic index (after a 14-hour nocodazoletreatment) in cells depleted of endogenousBubR1, and expressing BubR1-WT, BubR1-KEN26AAA, BubR1-KEN304AAA andBubR1-E413K. Values are mean ± s.e.m. ofthree independent experiments (180-250 cellsper experiment). (C)Representative stills fromtime-lapse videomicroscopy experimentsillustrating mitotic progression of HeLa cellsstably expressing histone H2B-GFP depleted ofendogenous BubR1 and rescued with mCherryempty vector or mCherry-BubR1-WT,mCherry-BubR1-KEN26AAA, mCherry-BubR1-KEN304AAA or mCherry-BubR1-E413K. Images were acquired at the indicatedtimepoints (hour: minutes) after the start ofchromosome condensation. White arrowindicates lagging chromosomes. (D)Box plotillustrating mitotic timing (chromosomecondensation – anaphase onset) in cells treatedas in C.*P<0.05, Student’s t-test. ‘n’ representsThe total number of cells (n) from threeindependent experiments are given above eachbox. (E)Bar graph illustrating the percentage ofcells in different mitotic stages (prophase-prometaphase, metaphase and anaphase) in cellsdepleted of endogenous BubR1 and rescuedwith MYC–BubR1-WT, MYC–BubR1-KEN26AAA, MYC–BubR1-KEN304AAA andMYC–BubR1-E413K, 11 hours after releasefrom thymidine block. Rescued cells wereidentified by MYC staining, and mitotic stagewas determined by staining for a-tubulin andDAPI. Values are mean ± s.e.m. of threeindependent experiments (n100-140 cells perexperiment).

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 8: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

91Uncoupling of BubR1 mitotic functions

WT, in line with their rapid and premature exit from mitosis. Bycontrast, lagging KTs were observed in BubR1-KEN304AAAanaphases at a much lower frequency than in the other SAC mutants(Fig. 6A,B). This suggests that the longer duration of mitoses inBubR1-KEN304AAA-expressing cells may allow sufficient timefor establishing accurate KT-MT attachments.

As with the BubR1 phosphorylation site mutants, we sought toinvestigate whether the motifs important for the SAC function ofBubR1 are also important for the KT-MT attachment function. Tothis end we treated cells again as shown in Fig. 2A and thenexamined metaphase alignment in the presence of MG132.Congression defects caused by endogenous BubR1 depletion werelargely rescued by BubR1-WT, -KEN26AAA, or -KEN304AAAexpression, but not by expression of BubR1-E413K (Fig. 6C).Together, these results indicate that whereas BubR1 KEN motifs

contribute to the SAC, these motifs are dispensable for chromosomeattachments. By contrast, the GLEBS motif of BubR1 is requiredfor both SAC and KT-MT attachment functions.

The Plk1-dependent BubR1 mitotic upshift correlates with itsfunction in maintaining stable KT-MT attachments (Elowe et al.,2007; Matsumura et al., 2007). We therefore tested whether BubR1KEN and GLEBS mutants also become upshifted in mitosis inresponse to nocodazole treatment. Whereas MYC–BubR1-WT, -KEN26AAA and -KEN304AAA mutants exhibited the double-bandpattern characteristic of BubR1, the mitotic upshift was no longerseen in MYC–BubR1-E413K (Fig. 6D). However, because theBubR1 upshift during mitosis is a result of Plk1 activity, it is notindicative of the phosphorylation sites described here. Thus, to studypossible effects of BubR1 mutations within the KEN and GLEBSmotifs on S670 phosphorylation we used the anti-pS670 antibody

Fig. 6. Chromosome congression andphosphorylation of BubR1 KEN-box and GLEBSdomain mutants. (A)HeLa cells were depleted ofendogenous BubR1 and rescued with MYC controlempty vector, MYC-tagged BubR1-WT, BubR1-KEN26AAA, BubR1-KEN304AAA and BubR1-E413K. Cells were then released from a thymidineblock for 11 hours before being fixed and stained withantibodies against MYC (red), CREST (green). DNAwas visualized with DAPI (blue) staining.Representative images of anaphases in cellsexpressing each of the BubR1 constructs are shown.Scale bar: 10m. (B)Bar graph illustratingpercentage of cells, treated as in A, initiating anaphaseeither fully aligned, with ≤2 lagging KTs, or >2lagging KTs. Values are mean ± s.e.m. of threeindependent experiments (n40-65 cells perexperiment). (C)Bar graph illustrating the percentageof cells in either prophase-prometaphase or aligned atmetaphase. Cells were depleted of endogenous BubR1and rescued as in A, released from a thymidine blockfor 10 hours, and arrested for 3 hours in MG132before being fixed. Cells were then stained with anti-MYC antibodies to identify rescued cells, and with a-tubulin antibodies and DAPI to facilitate identificationof the mitotic stage. (D)Electrophoretic mobility ofMYC-tagged BubR1-WT, -KEN26AAA, -KEN304AAA, and -E413K. MYC-tagged constructswere transfected into HeLa cells, arrested inthymidine for 24 hours, and subsequently releasedfrom this block into nocodazole for 16 hours beforemitotic cells were harvested by shake-off. Cell lysateswere resolved by SDS-PAGE and western blottingwith anti-MYC antibodies. a-Tubulin was used as aloading control (lower panel). (E)HeLa cells weretreated as in A. After fixation, cells were stained withanti-pS670 (red), and anti-MYC (green) antibodies.DNA was visualized using DAPI. Scale bar: 10m.(F)MYC-tagged BubR1-WT, -KEN26AAA, -KEN304AAA and -E413K were expressed in HeLacells. Cells were arrested in thymidine for 24 hours,and then released for 1 hour into nocodazole beforemitotic cells were harvested by shake-off. MYC-tagged proteins were subsequentlyimmunoprecipitated from total cells lysates, and thenresolved by SDS-PAGE and western blotting withanti-pS670, anti-pS676. Equal input is demonstratedby western blotting with anti-MYC antibodies.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 9: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

92

for immunofluorescence microscopy (Fig. 6E) and western blotting(Fig. 6F). As demonstrated by anti-pS670 antibody staining, BubR1-WT expression in HeLa cells depleted of endogenous BubR1resulted in phosphorylation of S670 and, similarly, robust pS670staining was detected at KTs in both BubR1-KEN26AAA- andBubR1-KEN304AAA-expressing cells (Fig. 6E). By contrast,BubR1-E413K expressing cells showed no anti-pS670 staining (Fig.6E). We considered the possibility that S670 phosphorylation wasnot detected in these cells because of a loss of BubR1-E413K fromthe KT and a corresponding dilution of the epitope into thecytoplasm. However, when the phosphorylation state of the BubR1mutants was examined by western blotting, we again observed thatBubR1-WT, BubR1-KEN26AAA and BubR1-KEN304AAAreadily became phosphorylated at both S670 and S676, but BubR1-E413K was not phosphorylated at either residue (Fig. 6F). Takentogether, these observations suggest that BubR1 phosphorylation,and thus BubR1 functionality during KT attachment, depends onthe BubR1-Bub3 interaction. This is lost in the BubR1-E413Kmutant, but not in either KEN-box mutant.

DiscussionHere, we demonstrate that the mitotic functions of BubR1 areregulated by distinct motifs and thus can be uncoupled from eachother (Fig. 7). Loss of BubR1 phosphorylation results in faulty KT-MT attachments and poor chromosome congression (Elowe et al.,2007; Huang et al., 2008; Matsumura et al., 2007). Furthermore,phosphorylation, at least at the sites described here, appears to belargely dispensable for the SAC functions of BubR1. By contrast,although KEN motifs are essential for SAC functionality of BubR1,no direct role was observed in either chromosome congression orBubR1 phosphorylation. Finally, the GLEBS domain is essentialfor both SAC activity and BubR1 phosphorylation, and thus foraccurate and timely chromosome congression and separation.

We have identified five proline-directed phosphorylation siteson BubR1. We demonstrate that at least three of the five sitesidentified are direct Cdk1 targets in vitro. Huang et al. recentlysuggested that Mps1 might be a major kinase for BubR1 in vivo(Huang et al., 2008). Although we also observe loss of BubR1phosphorylation upon Mps1 inhibition (data not shown), we havebeen unable to directly phosphorylate BubR1 by Mps1 and thusfavor the view that the reported result may represent an indirecteffect, perhaps reflecting mislocalization of BubR1. Indeed, amislocalization of endogenous BubR1 upon Mps1 inhibition (ourunpublished results), and the lack of any detectable phosphorylationon the mislocalized BubR1-E413K mutant support the conclusionthat KT localization of BubR1 is required for its phosphorylation.Importantly, phosphorylation on BubR1 at one of these SP sites,S670, is significantly enhanced at unattached KTs but not at KTslacking tension, in excellent agreement with previous work (Huanget al., 2008).

Recent studies indicate that Cdk1-cyclin B1 localizes to KTsduring prometaphase, where it contributes to the correct attachmentof MTs to KTs and efficient chromosome alignment throughphosphorylation of local substrates (Bentley et al., 2007; Chen etal., 2008). Moreover, KT recruitment of Cdk1 is increased innocodazole- but not Taxol-treated cells (Bentley et al., 2007), inagreement with the increase in S670 phosphorylation observed hereupon nocodazole but not Taxol treatment.

Studies of yeast Mad3 and N-terminal fragments of murineBubR1, as well as our own observations reported here, indicate thatboth KEN motifs are required for SAC function (Burton and

Journal of Cell Science 123 (1)

Solomon, 2007; King et al., 2007; Malureanu et al., 2009;Sczaniecka et al., 2008). Importantly, however, our binding studiesreveal that the interaction between BubR1 and Cdc20 is in itselfnot sufficient for APC/CCdc20 inhibition, as full-length BubR1mutated at its KEN boxes bound Cdc20 efficiently, and yet cellsexpressing these mutants exited mitosis prematurely and could notarrest upon microtubule depolymerization. Nevertheless, we alsodemonstrate that efficient recruitment of Cdc20 is necessary forbinding and inhibition of the APC/C and that this strictly requiresthe C-terminal region of BubR1. It is possible that APC/C inhibitionprovided by the BubR1 KEN-box interaction with Cdc20 issufficient for cell survival during normal passage through mitosis,but that the C-terminal BubR1 Cdc20 binding region becomesessential for full SAC activity upon microtubule depolymerization.In support of this view, a mutant of murine BubR1 [mBubR1(525-700)], which lacks only the C-terminal Cdc20 docking site, supportscell survival but is unable to fully sustain the SAC (Malureanu etal., 2009).

Our data also suggest that the two KEN motifs make distinctcontributions to BubR1 functionality during mitosis. We found thatan N-terminal but not a C-terminal KEN-box peptide interactsdirectly with Cdc20, in agreement with data from both S. cerevisiaeand S. pombe (Burton and Solomon, 2007; King et al., 2007;Sczaniecka et al., 2008). This data supports the idea that BubR1can act as a competitive inhibitor of substrate binding through a

Fig. 7. Spindle checkpoint and chromosome congression functions ofBubR1 are separable. (A)WT BubR1 is phosphorylated in mitosis andlocalizes to the KT; it functions in both chromosome alignment and the SAC.(B)Expression of non-phosphorylatable BubR1 supports the SAC but resultsin severe KT-MT attachment defects. (C)Mutation of the KEN-box motifs orreduction of Cdc20 binding abrogates the SAC but not chromosomecongression. (D)Mutation of the GLEBS domain abrogates BubR1phosphorylation and impairs both chromosome congression and SAC function.(E)The contribution of the different BubR1 domains to the SAC andchromosome congression. Dashed lines indicate results from Huang et al.(Huang et al., 2008).

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 10: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

93Uncoupling of BubR1 mitotic functions

direct interaction between the N-terminal KEN box and Cdc20. Inaddition, the KEN26-Cdc20 interaction may be required for therecently reported Cdc20 turnover in early mitosis, which is thoughtto be required for maintaining the SAC (Nilsson et al., 2008; Kinget al., 2007). It is attractive to speculate that the N-terminal KENbox of BubR1 plays a dual role in the SAC: first, by establishingtight binding to Cdc20 it may exclude bona fide mitotic substrates,and second, by functioning as a destruction box in trans, it mayfacilitate Cdc20 ubiquitylation and degradation.

The role of the C-terminal KEN box is less clear. Although theKEN304 motif is required for SAC function, cells expressingBubR1-KEN304AAA were consistently and significantly slowerin completing mitosis and exited with a significantly lower incidenceof lagging KTs than cells expressing other SAC mutants. This subtledifference would not have been readily detected in end point survivalassays such as those used in the studies on yeast Mad3 and murineBubR1 (Burton and Solomon, 2007; King et al., 2007; Malureanuet al., 2009; Sczaniecka et al., 2008). One possible function of theC-terminal KEN box could relate to the proper docking andorientation of BubR1 on the APC/C, similar to what has beenproposed for the D-box of Emi1 (Miller et al., 2006). In this context,it is interesting that Drosophila BubR1 has only one KEN box,corresponding to the N-terminal KEN motif of the vertebrateenzyme. How Drosophila compensates for the absence of a C-terminal KEN box is not presently clear.

Several lines of evidence indicate that BubR1-Bub3 associationis important for SAC function and accurate chromosomecongression. First, Bub3 is one of the original SAC componentsidentified in S. cerevisiae (Hoyt et al., 1991), and loss of Bub3expression or the Mad3p-Bub3p interaction confers benomylsensitivity (Hardwick et al., 2000). Our own results corroborate thedata from S. cerevisiae and demonstrate that cells expressingBubR1-E413K, a mutant that cannot bind Bub3 or localize to KTs,have a defective SAC (Fig. 5B). Second, BubR1 dynamicallyassociates with KTs, and FRAP studies show that BubR1 and Cdc20display similar biphasic kinetics at unattached KTs, arguing thatthey shuttle off the KT in a complex (Howell et al., 2004). Third,overexpression of a peptide encompassing the BubR1 GLEBS motifwas shown to disrupt the nocodazole-activated SAC in HeLa cells(Harris et al., 2005). Taken together, these data strongly supportthe view that the GLEBS motif in BubR1 contributes to the SACfunction of BubR1. In addition, we show here that BubR1-E413Kdoes not exhibit the characteristic upshift in mitosis and does notbecome phosphorylated at either S670 or S676, two sites shown tobe critical for productive KT-MT attachments. Whether the loss offunctionality of the BubR1 GLEBS mutant reflects an impairedrecruitment of BubR1 to KTs or a disruption of the BubR1-Bub3interaction (or both) remains to be clarified.

Recent studies were interpreted to suggest that cytosolic BubR1is sufficient for efficient mitosis. First, Drosophila cid (CENP-A)mutants retain an intact SAC response to spindle disruption despitethe inability of SAC components, including BubR1, to target toKTs (Blower et al., 2006). Second, a cytosolic murine BubR1fragment was recently reported to support cell survival (Malureanuet al., 2009). However, although unattached KTs are apparently notstrictly required for MCC formation in vitro, they do accelerate thisprocess (Kulukian et al., 2009). In vivo, the rate at which MCCformation occurs may not be sufficient to support a fully functionalcheckpoint. Anchoring of BubR1 to the KT through Bub3 wouldprovide an elevated concentration of BubR1 readily available forassociation with the primed Cdc20-Mad2 complexes, and this may

well be important for timely MCC formation. Although it is possiblethat the requirement for the GLEBS domain reflects an essentialfunction of the BubR1-Bub3 complex that is independent of KTlocalization, it remains difficult to rationalize how a cytoplasmicBubR1 fragment that lacks not only the GLEBS domain but alsophosphorylation sites known to be important for chromosomecapture and alignment (Elowe et al., 2007; Huang et al., 2008;Matsumura et al., 2007) would be able to confer stable KT-MTinteractions (Malureanu et al., 2009).

Materials and MethodsCell culture, synchronization, transfections and siRNA depletionHeLa S3, HEK293T and HeLa S3 cells expressing histone H2B-GFP were routinelymaintained in DMEM (Invitrogen) supplemented with 10% FBS and penicillin-streptomycin (100 IU/ml and 100 mg/ml, respectively, Gibco). MG132 (Calbiochem)was used at 20 M for 3 hours unless otherwise stated. For synchronization studies,nocodazole and thymidine were used at 300 nM and 2 mM respectively for 16 hoursunless otherwise stated. All BubR1 constructs were generated in the pcDNA3.1plasmid (Invitrogen), driven by the CMV promoter, and modified to carry an N-terminal triple-MYC tag.

Kinase assaysIn vitro phosphorylation of recombinant BubR1 was carried out in 30 l of kinasereaction buffer as previously described (Elowe et al., 2007). Recombinant active Cdk1was purchased from Upstate Biotechnology, and GST-Mps1 from Invitrogen. ForCdk1 assays on peptides immobilized on cellulose membranes, dried membraneswere first washed in ethanol and then hydrated in kinase buffer [50 mM Tris-HClpH 7.5, 10 mM MgCl2, 1 mM DTT (dithiothreitol), 100 M NaF, 10 M sodiumvanadate] for 1 hour, followed by overnight blocking in kinase buffer with 100 mMNaCl and 0.5 mg/ml BSA. The next day, the membrane was blocked again withkinase buffer containing 1 mg/ml BSA, 100 mM NaCl and 50 M non-radioactiveATP at 30°C for 45 minutes. The blocking solution was subsequently replaced withkinase reaction buffer containing 0.2 mg/ml BSA, 50 Ci/ml [g-32P]ATP (3000Ci/mmol, 10 mCi/ml), 2 g/ml recombinant Cdk1, and 50 M ATP for 3 hours ona shaker at 30°C. The membranes were then washed extensively: 10�15 minutes in1 M NaCl, 3�5 minutes in H2O, 3�15 minutes 5% H3PO4, 3�5 minutes in H2O,and then sonicated overnight in 8 M urea, 1% SDS (w/v) and 0.5% (v/v) -mercaptoethanol to remove residual nonspecific radioactivity. Phosphorylation wasvisualized by autoradiography.

Immunofluorescence and time-lapse microscopyCells grown on coverslips were fixed and permeabilized simultaneously in PTEMFbuffer (0.2% Triton X-100, 20 mM PIPES pH 6.8, 1 mM MgCl2, 10 mM EGTA and4% formaldehyde). Processing for immunofluorescence and image acquisition on aDeltavision microscope (Applied Precision) were performed as previously described.For time-lapse microscopy, all treatments within a single experiment were performedsimultaneously. During imaging, the atmosphere was maintained at a temperature of37°C, humidity 60% and 5% CO2. Imaging was performed using a Zeiss AxioObserver Z1 microscope equipped with a Plan Neofluar 40� objective. Metamorph7.1 software (Molecular Devices) was used to collect and process data. Images werecaptured at 3-minute intervals for 16 hours.

Antibodies and antibody productionThe monoclonal BubR1 antibody was previously described (Elowe et al., 2007). Anti-p55Cdc20 (Santa Cruz Biotechnology), anti-Bub3 (BD Transduction labs), anti-MYC(9E10, ATCC), anti-a-tubulin (DM1A, Sigma), anti-Cdc27 (BD Transduction labs),anti-APC7 (Biolegend), anti-APC4 (Santa Cruz Biotechnology), anti-Bub1(Chemicon), as well as CREST anti-human auto-immune serum (Immunovision),were obtained commercially. Anti-pS670 polyclonal antibody was generated byimmunizing rabbits with KLH-conjugated phosphopeptide (H-CSIKKLS(P)PIIED-OH), and then isolated from a protein-A-purified IgG fraction using the same peptide.For immunofluorescence experiments, all primary antibodies were detected withCy2/Cy3-conjugated donkey antibodies (Dianova) and Alexa-Fluor-647-conjugatedgoat antibodies (Invitrogen). For western blots, signals were detected using HRP-conjugated anti-mouse or anti-rabbit antibodies (Pierce).

Peptide array synthesis and spots blottingPeptide arrays were constructed according to the Spots-synthesis method accordingthe manufacturer’s directions (Intavis). For Cdc20 binding experiments, purified GST-Cdc20 fusion protein generated in SF9 insect cells was added at 5 g/ml in TBSTand incubated together with the membrane overnight at 4°C. Membranes were washedthree times in TBST and bound protein was visualized with anti-Cdc20 antibodies.The sequences of synthesized peptides are as follows: KEN30: DEWELSKEN -VQPLRQ; KEN304: PPMPRAKENELQAGP; S543: SEKKNKSPPADP; S574:TSNEDVSPDVCD; S670: LSIKKLSPIIED; S720: SENPTQSPWCSQ; S1043:KVGKLTSPGALL.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence

Page 11: Uncoupling of the spindle-checkpoint and chromosome … · 2010-01-12 · Uncoupling of BubR1 mitotic functions 85 for cell survival, whereas mutation of either KEN box led to loss

94 Journal of Cell Science 123 (1)

SILAC labeling with L-[6-13C, 4-15N]arginine and L-[6-13C, 2-15N]lysineHeLa S3 cells were cultured in DMEM formulated with either unlabeled L-lysine orL-arginine or labeled with L-[6-13C, 4-15N]arginine and L-[6-13C, 2-15N]lysine(Cambridge Isotope Laboratories) at a concentration of 44 and 86 g/ml respectively,and supplemented with 10% dialyzed fetal bovine serum. Extracts prepared from4�107 unlabelled nocodazole-arrested cells and of 4�107 isotopically labeledthymidine-blocked cells were mixed at a ratio of 1:1. This mixture was divided intoequal parts, and immunoprecipitation was performed with anti-BubR1 monoclonalantibody or 9E10 anti-MYC monoclonal antibody as a negative control. Sampleswere separated by SDS-PAGE and excised gel fragments were processed for massspectrometry.

The authors would like to thank Alessandro Tosi and Claudia Szalmafor technical assistance, Roman Körner for assistance with massspectrometry, and Lily Wang and Anna Santamaria for critical readingof the manuscript and insightful discussions. S.E. holds a post-doctoralfellowship from the Canadian Institute of Health Research (CIHR). Thiswork was supported by the Max Planck Society.

Supplementary material available online athttp://jcs.biologists.org/cgi/content/full/123/1/84/DC1

ReferencesBentley, A. M., Normand, G., Hoyt, J. and King, R. W. (2007). Distinct sequence elements

of cyclin B1 promote localization to chromatin, centrosomes, and kinetochores duringmitosis. Mol. Biol. Cell 18, 4847-4858.

Blower, M. D., Daigle, T., Kaufman, T. and Karpen, G. H. (2006). Drosophila CENP-A mutations cause a BubR1-dependent early mitotic delay without normal localizationof kinetochore components. PLoS Genet. 2, e110.

Burton, J. L. and Solomon, M. J. (2007). Mad3p, a pseudosubstrate inhibitor of APCCdc20in the spindle assembly checkpoint. Genes Dev. 21, 655-667.

Chen, Q., Zhang, X., Jiang, Q., Clarke, P. R. and Zhang, C. (2008). Cyclin B1 is localizedto unattached kinetochores and contributes to efficient microtubule attachment and properchromosome alignment during mitosis. Cell Res. 18, 268-280.

Davenport, J., Harris, L. D. and Goorha, R. (2006). Spindle checkpoint function requiresMad2-dependent Cdc20 binding to the Mad3 homology domain of BubR1. Exp. CellRes. 312, 1831-1842.

Ditchfield, C., Johnson, V. L., Tighe, A., Ellston, R., Haworth, C., Johnson, T., Mortlock,A., Keen, N. and Taylor, S. S. (2003). Aurora B couples chromosome alignment withanaphase by targeting BubR1, Mad2, and Cenp-E to kinetochores. J. Cell Biol. 161,267-280.

Elowe, S., Hummer, S., Uldschmid, A., Li, X. and Nigg, E. A. (2007). Tension-sensitivePlk1 phosphorylation on BubR1 regulates the stability of kinetochore microtubuleinteractions. Genes Dev. 21, 2205-2219.

Fang, G. (2002). Checkpoint protein BubR1 acts synergistically with Mad2 to inhibitanaphase-promoting complex. Mol. Biol. Cell 13, 755-766.

Hardwick, K. G., Johnston, R. C., Smith, D. L. and Murray, A. W. (2000). MAD3encodes a novel component of the spindle checkpoint which interacts with Bub3p,Cdc20p, and Mad2p. J. Cell Biol. 148, 871-882.

Harris, L., Davenport, J., Neale, G. and Goorha, R. (2005). The mitotic checkpoint geneBubR1 has two distinct functions in mitosis. Exp Cell Res 308, 85-100.

Howell, B. J., Moree, B., Farrar, E. M., Stewart, S., Fang, G. and Salmon, E. D. (2004).Spindle checkpoint protein dynamics at kinetochores in living cells. Curr. Biol. 14, 953-964.

Hoyt, M. A., Totis, L. and Roberts, B. T. (1991). S. cerevisiae genes required for cellcycle arrest in response to loss of microtubule function. Cell 66, 507-517.

Huang, H., Hittle, J., Zappacosta, F., Annan, R. S., Hershko, A. and Yen, T. J. (2008).Phosphorylation sites in BubR1 that regulate kinetochore attachment, tension, and mitoticexit. J. Cell Biol. 183, 667-680.

Hwang, L. H., Lau, L. F., Smith, D. L., Mistrot, C. A., Hardwick, K. G., Hwang, E.S., Amon, A. and Murray, A. W. (1998). Budding yeast Cdc20: a target of the spindlecheckpoint. Science 279, 1041-1044.

King, E. M., van der Sar, S. J. and Hardwick, K. G. (2007). Mad3 KEN boxes mediateboth Cdc20 and Mad3 turnover, and are critical for the spindle checkpoint. PLoS ONE2, e342.

Kiyomitsu, T., Obuse, C. and Yanagida, M. (2007). Human Blinkin/AF15q14 is requiredfor chromosome alignment and the mitotic checkpoint through direct interaction withBub1 and BubR1. Dev. Cell 13, 663-676.

Kulukian, A., Han, J. S. and Cleveland, D. W. (2009). Unattached kinetochores catalyzeproduction of an anaphase inhibitor that requires a Mad2 template to prime Cdc20 forBubR1 binding. Dev. Cell 16, 105-117.

Lampson, M. A. and Kapoor, T. M. (2005). The human mitotic checkpoint protein BubR1regulates chromosome-spindle attachments. Nat. Cell Biol. 7, 93-98.

Malureanu, L. A., Jeganathan, K. B., Hamada, M., Wasilewski, L., Davenport, J. andvan Deursen, J. M. (2009). BubR1 N terminus acts as a soluble inhibitor of cyclin Bdegradation by APC/C(Cdc20) in interphase. Dev. Cell 16, 118-131.

Matsumura, S., Toyoshima, F. and Nishida, E. (2007). Polo-like kinase 1 facilitateschromosome alignment during prometaphase through BubR1. J. Biol. Chem. 282, 15217-15227.

Miller, J. J., Summers, M. K., Hansen, D. V., Nachury, M. V., Lehman, N. L., Loktev,A. and Jackson, P. K. (2006). Emi1 stably binds and inhibits the anaphase-promotingcomplex/cyclosome as a pseudosubstrate inhibitor. Genes Dev. 20, 2410-2420.

Musacchio, A. and Salmon, E. D. (2007). The spindle-assembly checkpoint in space andtime. Nat. Rev. Mol. Cell. Biol. 8, 379-393.

Nilsson, J., Yekezare, M., Minshull, J. and Pines, J. (2008). The APC/C maintains thespindle assembly checkpoint by targeting Cdc20 for destruction. Nat. Cell Biol. 10, 1411-1420.

Sczaniecka, M., Feoktistova, A., May, K. M., Chen, J. S., Blyth, J., Gould, K. L. andHardwick, K. G. (2008). The spindle checkpoint functions of Mad3 and Mad2 dependon a Mad3 KEN box-mediated interaction with Cdc20-anaphase-promoting complex(APC/C). J. Biol. Chem. 283, 23039-23047.

Tang, Z., Bharadwaj, R., Li, B. and Yu, H. (2001). Mad2-Independent inhibition ofAPCCdc20 by the mitotic checkpoint protein BubR1. Dev. Cell 1, 227-237.

Yu, H. (2002). Regulation of APC-Cdc20 by the spindle checkpoint. Curr. Opin. Cell Biol.14, 706-714.

Jour

nal o

f Cel

l Sci

ence

Jour

nal o

f Cel

l Sci

ence