Main

Cdk1–cyclin B phosphorylation of a conserved site in the carboxy terminus of PP1 depresses PP1 activity at mitotic commitment1,7,8,9,10. Declining Cdk1–cyclin B levels then allow the compromised PP1 to dephosphorylate itself to promote a return to full activity1,7. Of the two fission yeast PP1 enzymes, PP1Sds21 and PP1Dis2, only PP1Dis2 harbours the conserved inhibitory phosphorylation site2,9 (Extended Data Fig. 1a). In vitro PP1Dis2 activity assays recapitulated previous observations that T316 phosphorylation by Cdk1–cyclin B depressed activity (Fig. 1a and Extended Data Fig. 1b, c)1,7,9. Mutating T316 to aspartic acid to mimic phosphorylation reduced the activity to a similar degree to phosphorylation by Cdk1–cyclin B (Fig. 1a and Extended Data Fig. 1c). Replacement of PP1dis2+ with a PP1dis2.T316A allele increased PP1Dis2 levels, whereas they were reduced in PP1dis2.T316D (Fig. 1b), indicating that phosphorylated T316 might act as a phospho-degron. Since this interpretation conflicted with reports of stable PP1Dis2 levels throughout mitosis10,13, we monitored PP1Dis2 levels with both low and high antibody dilutions as size-selected cells synchronously transited the cell cycle. A transient reduction in PP1Dis2 levels as T316 phosphorylation peaked (Fig. 1c) was blocked when proteosome function was inhibited (Extended Data Fig. 1e). Consistently, PP1Dis2 levels were persistently low in PP1dis2.T316D and persistently high in PP1dis2.T316A (Fig. 1c and Extended Data Fig. 1f, g), indicating that phosphorylation of T316 by Cdk1–cyclin B both reduces PP1Dis2 levels and inhibits its phosphatase activity.

Figure 1: PP1Dis2 T316 phosphorylation and stability.
figure 1

a, EnzChek phosphatase assays (Extended Data Fig. 1b) of PP1Dis2 isolated from the indicated strains by antibody precipitation. ‘+Cdk1–cyclin B’ indicates the addition of sepharose beads to which covalently linked p13Suc1 protein had recruited Cdk1–cyclin B from S. pombe cell extracts. NIPP1 is a highly specific PP1 inhibitor. Purvalanol (Pur.) is a Cdk1 inhibitor. b, c, PP1Dis2 and T316 phosphorylation (T316Phos) levels (Rpn12mts3.1 is a temperature-sensitive proteasome mutation). For b, n = 5 biological replicates; error bars show standard deviation (s.d.). c, Size-selected cells transit the cell division cycle. The septation profile is for the wild-type culture. n = 2 biological with two technical replicates each. PP1Dis2.T316D and PP1Dis2.T316A levels from the experiments in Extended Data Fig.1f, g are superimposed on this wild-type data set.

PowerPoint slide

PP2A holoenzymes combine a catalytic and scaffolding subunit with one of four regulatory B subunits14, of which PP2A-B55 and PP2A-B56 have been linked to mitotic control3,4,5,15,16. We noticed that the B55 and B56 regulatory subunits had highly conserved PP1-docking-site consensus motifs (RVxF/RxVxF)12 (Fig. 2a). The Schizosaccharomyces pombe genome encodes one B55 (B55Pab1) and two B56 subunits (B56Par1 and B56Par2)17,18, and we found that both B55Pab1 and B56Par1 associated with PP1Dis2 in immunoprecipitation assays (Fig. 2b and Extended Data Fig. 2a–d). B56Par1 also bound PP1Dis2 in a yeast two-hybrid assay (Extended Data Fig. 3), and the functional replacement of the PP1-docking site of the morphogenesis regulator Wsh3 (also known as Tea4)19 by the SKEVLF motif of B56Par1 confirmed its ability to recruit PP1 (Extended Data Fig. 4a–d). The interaction between PP1Dis2 and B55Pab1 was abolished by mutating the PP1-docking consensus motif (Fig. 2b and Extended Data Fig. 2a). No association was found between PP1Sds21 and any regulatory subunit (Extended Data Fig. 2b, c), nor between B56Par2 and PP1Dis2 (Extended Data Figs 2d and 3). L482 of B56Par2 occupies a position occupied by only valine or isoleucine in validated PP1-docking sites12. Changing this leucine to valine allowed PP1Dis2 to bind B56Par2 (Extended Data Fig. 2e).

Figure 2: PP1Dis2 recruitment to PP2A-B55Pab1 and PP2A-B56Par1 regulates chromosome segregation.
figure 2

a, Clustal W alignments of PP1-docking motifs in B55 and B56 molecules. Consensus residues are in bold. b, f, g, Immunoprecipitation (IP) reactions probed with PP1Dis2(ref. 20) and T316Phos polyclonal and 12CA5 and 366 monoclonal (HA and Pk epitopes, respectively) antibodies. Asynch., asynchronous. Asterisk indicates PP1dis2.T316A. f, Mitotic progression was scored by tubulin staining with telophase defined by post-anaphase arrays of microtubules. b, n = 6; error bars show s.d. c, d, 4′,6-Diamidino-2-phenylindole (DAPI) staining. e, Tubulin immunofluorescence staining for Cdk1cdc2.33 ‘arrest/release’ experiments scored mitotic progression. Biological replicates: for b, n = 6; for eg, n = 2.

PowerPoint slide

Core K/RxVxF PP1-docking motifs can be accompanied by secondary motifs12. Although the B55Pab1 docking site is an isolated K/RxVxF motif, the GLLR sequence of B56Par1 bears a striking resemblance to the secondary element G/SILK/R11,12 (Fig. 2a, green box). Mutating the GLLR motif (B56par1.G367V) mimicked mutating the R/KxVxF motif (B56par1.K379AV381AF383A) in abolishing interactions between PP1Dis2 and B56Par1 in two-hybrid assays (Extended Data Fig. 3), and in compromising their ability to co-immunoprecipitate (Fig. 2b and Extended Data Fig. 2a). Mutating both motifs had an additive effect, severely compromising the affinity of B56Par1 for PP1Dis2 in co-immunoprecipitation assays (Fig. 2b and Extended Data Fig. 2a). Finally, the GLLR motif efficiently substituted for the characterized PP1-binding GILK motif of Cut12 (Extended Data Fig. 4e–g)20. We conclude that GLLR is a ‘G/SILK/R’ motif.

To confirm that the motifs we identified represent genuine PP1Dis2-docking sites, we generated PP1-docking-site-null mutants (PDSN) of B55pab1 (R52A/V54A/F56A) and B56par1 (G367V/K379A/V381A/F383A), and purified the respective holoenzymes and wild-type controls from yeast cultures. The purified proteins were mixed with purified PP1Dis2 and the PP1Dis2 re-isolated to identify interacting partners. In each case, wild-type but not PDSN holoenzymes were captured by PP1Dis2 (Extended Data Fig. 5a–d). Moreover, PP1Dis2 co-migrated with the largest form of each of the wild-type PP2A holoenzyme complexes in size-exclusion chromatography (Extended Data Fig. 5e). We conclude that both B56Par1 and B55Pab1 contain genuine PP1Dis2-docking motifs.

To assess the mitotic defects of the B55pab1.PDSN and B56par1.PDSN PP1-docking site mutants (Fig. 2c, d), we exploited transient arrest at the G2/M boundary with the temperature-sensitive Cdk1cdc2.33 mutation to synchronize mitotic progression21 (Fig. 2e). Blocking PP1Dis2 recruitment to either B55Pab1 or B56Par1 in synchronized divisions generated major errors in chromosome segregation and delayed the metaphase/anaphase transition (Fig. 2e and Extended Data Fig. 6a). To assay the timing of PP1Dis2 association with each PP2A holoenzyme, we used a Cdk1cdc2.33 strain in which B55Pab1 and B56Par1 were fused to different epitope tags, SV5 P protein (Pk) and haemagglutinin (HA) (Cdk1cdc2.33, B55pab1.Pk and B56par1.HA). PP1Dis2 levels in B55Pab1.Pk precipitates peaked at metaphase (25–35 min) whereas PP1Dis2 levels in B56Par1.HA immunoprecipitates peaked later as spindles disassembled in telophase (60–70 min) (Fig. 2f).

An established assay for PP2A-B56Par1 (ref. 22), in which PP1Dis2 displayed no activity (Extended Data Fig. 6b–d), revealed a mitotic decline in bulk PP2A-B56Par1 activity before recovery at the start of the next cycle (Fig. 3a). PP2A-B56Par1 activity was severely compromised when either the GLLR (GILK) or KEVLF (RxVxF) motifs were individually mutated (Extended Data Fig. 6e, f), and abolished when both were simultaneously mutated to disrupt PP1Dis2 recruitment (Fig. 3b and Extended Data Fig. 6g). This indicated that PP1Dis2 could activate PP2A-B56Par1; we confirmed this by adding immunopurified PP1Dis2 to PP2A-B56Par1 that had been isolated by immunoprecipitation from synchronized cells that lacked PP1Dis2 (PP1dis2) (Fig. 3c). Moreover, PP1Dis2 activated wild-type PP2A-B56Par1 but not the PP1-docking-site mutant (Fig. 3c, d and Extended Data Fig. 6g, h), and genetically inhibited PP1Dis2.T316D was unable to activate PP2A-B56Par1 (Fig. 3c and Extended Data Fig. 6h). Similar docking-site-dependent activity enhancement of purified PP2A-B56Par1 enzymes by purified PP1Dis2 (Extended Data Fig. 5f) further confirmed that recruitment of active PP1Dis2 to the PP2A-B56Par1 docking site reactivated PP2A-B56Par1.

Figure 3: Phosphorylation status of S378 determines competence to recruit active PP1Dis2 to PP2A-B56Par1 to promote PP2A-B56Par1 activity.
figure 3

a, b, gi, Assays of PP2A-B56Par1 activity after B56Par1.HA immunoprecipitation (with 12CA5 antibodies) of Cdk1cdc2.33 arrest/release synchronized cultures followed for one (b, gi) or two (a) cycles. Activity changes (loss of GST–Rec8391–561 32P radioactivity22) at each point were normalized to the activity of B56Par1.HA from an asynchronous (Asynch.) culture processed in parallel on the same gel (penultimate lane). sub, phosphorylated substrate. c, d, Aliquots of PP1Dis2 protein immunoprecipitates (isolated under high salt (1.2 M NaCl) extraction conditions to disassociate partners) from Cdk1cdc2.33 arrested (interphase) cultures were added to PP2A-B56Par1 assays from Cdk1cdc2.33 dis2.Δ cultures (Extended Data Fig. 6). e, f, Co-immunoprecipitation assays as for Fig. 2b. Asterisks indicate extracts from asynchronous B56par1.HA cultures. j, Phenotype analysis of Cdk1cdc2.33 synchronized mitoses as for Fig. 2e. Biological replicates: for a, n = 3; for all other panels, n = 2.

PowerPoint slide

Phosphorylation between the G/SILK/R and RxVxF motifs of a bipartite docking site can block PP1 recruitment20. Mass spectrometry analysis of B56Par1 from mitotic cells appeared to indicate phosphorylation on S378 between the GLLR and KEVLF motifs (boxed residue in Fig. 2a; data not shown). Attempts to generate antibodies to recognize B56Par1 when phosphorylated on S378 failed, prompting us to generate a polyclonal antibody that would recognize B56Par1 when phosphorylated on both S377 and S378 (Extended Data Fig. 7a). This antibody (B56-Phos) revealed increasing phosphorylation in mitosis that declined as PP1Dis2 was recruited to PP2A-B56Par1 ( Fig. 4a). Mutating S378 to alanine to block phosphorylation (B56par1.S378A) promoted persistent PP1Dis2 recruitment to PP2A-B56Par1 and persistently high levels of PP2A-B56Par1 activity throughout mitosis that depended upon the presence of PP1Dis2 (Fig. 3e, g, h and Extended Data Fig. 6i, j). Conversely, when S378 was mutated to aspartic acid to mimic phosphorylation (B56par1.S378D), this severely compromised PP1Dis2 recruitment and B56Par1.S378D activity remained low throughout division (Fig. 3f, i and Extended Data Fig. 6k). Anaphase was delayed and frequently abnormal in B56par1.S378D cells (Fig. 3j), and it was striking that the persistent mitotic association of PP1Dis2 with PP2A-B56Par1 in B56par1.S378A cells generated similar mitotic errors (Fig. 3j). Thus, dynamic phosphate turnover on S378 appears to be crucial for orderly mitotic progression. As anticipated from the match to the consensus phosphorylation site for the human polo-like kinase PLK1 (N/D/ExS/T)23, PoloPlo1 was solely responsible for S378 phosphorylation (Fig. 4b, c).

Figure 4: PP2A-B55Pab1 dephosphorylation of B56Par1 promotes PP1Dis2 recruitment to activate PP2A-B56Par1 phosphatase in a mitotic phosphatase relay.
figure 4

a, ei, Cdk1cdc2.33 synchronized mitoses. ac, e, B56-Phos antibodies (Extended Data Fig. 7a) detected phosphorylation on B56Par1 after immunoprecipitation from the indicated strains and the indicated treatments. PP1Dis2 levels were monitored where indicated. λ phosph., λ phosphatase; 3BrB-PP1, 3-[(3-bromophenyl)methyl]-1-tert-butylpyrazolo[3,4-d]pyrimidin-4-amine; PoloPlo1.KDHA, HA-tagged catalytically inactive; inhib., inhibitor. d, B56Par1 precipitates probed for PP1Dis2. fi, PP2A-B55Pab1 phosphatase assays (Extended Data Fig. 7c–f). j, Bulk activities are illustrated: local activities at any given time will depend on the specific balance of each activity at a particular site. (1) Cdk1–cyclin B activation represses all three phosphatase activities (Extended Data Fig. 9a). Direct phosphorylation of PP1Dis2 TPRR motif inhibits catalytic activity1,7,8,9,10 and promotes destruction. PP1Dis2 binds B55Pab1 but cannot reactivate PP2A-B55Pab1 while TPRR phosphorylation persists. PoloPlo1 phosphorylation of B56Par1 prevents PP1Dis2 recruitment to PP2A-B56Par1. (2) Declining Cdk1–cyclin B activity facilitates PP1 auto-reactivation. (3) Re-activated PP1Dis2 promotes PP2A-B55Pab1 reactivation. (4) PP2A-B55Pab1 dephosphorylates the PP1-docking site in B56Par1 less efficiently than PoloPlo1 phosphorylates it, keeping PP2A-B56Par1 activity low. (5) Declining PoloPlo1 activity enables PP2A-B55Pab1 to dephosphorylate B56Par1 and promote reactivating recruitment of PP1Dis2 (Extended Data Fig. 8). k, Flag-tagged B56γ and B56δ isoforms were stably expressed in HeLa-FRT cell lines, immunoprecipitated from mitotic cells isolated by shake-off from nocodazole-treated cells, and the immunoprecipitates probed with an antibody that recognizes all forms of PP1. Biological replicates: for k, n = 3; for all other panels, n = 2.

PowerPoint slide

PP1Dis2 recruitment to B56Par1 at telophase (Fig. 2f) indicated that S378 should be removed at this time, and the inability of B55pab1.Δ cells to recruit PP1Dis2 to B56Par1 (Fig. 4d) indicated that PP2A-B55Pab1 could be responsible for this dephosphorylation. In support of this, PP2A-B55Pab1 removed B56-Phos reactivity from B56Par1 in an in vitro assay (Extended Data Fig. 7b). This phosphatase activity peaked during mitosis and was abolished when PP1Dis2 recruitment to B55Pab1 was blocked by removal of PP1Dis2, when the PP1-docking site in B55Pab1 was ablated, and when genetically inhibited PP1dis2.T316D was recruited (Fig. 4f–i and Extended Data Fig. 7c–f). Consistently, the failure of PP1Dis2 to associate with PP2A-B56Par1 in B55pab1.Δ cells correlated with persistent phosphorylation on S378 throughout mitosis (B55pab1; Fig. 4e). Finally, purified PP1Dis2 enhanced the B56-Phos activity of purified PP2A-B55Pab1 in a docking-site-dependent manner (Extended Data Fig. 5f).

In summary, PP1Dis2, PP2A-B55Pab1 and PP2A-B56Par1 are linked in a phosphatase relay (Fig. 4j and Extended Data Fig. 8). At mitotic commitment all three activities are repressed4 (Extended Data Fig. 9a); subsequently PP1Dis2 activity recovers auto-catalytically as cyclin B is degraded. The binding of repressed, T316-phosphorylated PP1Dis2 to PP2A-B55Pab1 (Fig. 2g) promotes rapid restoration of PP2A -B55Pab1 activity upon reduction of Cdk1–cyclin B activity. Reactivated PP2A-B55Pab1 can then begin to dephosphorylate S378 of B56Par1 but is antagonized by PoloPlo1activity towards this site. Consequently, PP1Dis2 can only be recruited to PP2A-B56Par1 at telophase when PoloPlo1 activity declines (Fig. 4j and Extended Data Fig. 8). Thus, Cdk1–cyclin B phosphorylation of PP1 at mitotic commitment1,7,8,9,10 locks all three phosphatases in the ‘off’ state. This lock is bolted through a Cdk1–cyclin B phosphorylation-mediated reduction of PP1Dis2 levels. These controls join a range of other modes of PP1 and PP2A regulation, including Greatwall/ENSA control of PP2A-B55δ, and PP2A-B56 regulation by Bod1, to provide the accurate control of phosphorylation status that is critical for faithful cell division1,6,11,24,25.

The conservation of the PP1-docking-site motifs in PP2A regulatory subunits (Fig. 2a) prompted us to investigate the association between human PP1 and PP2A enzymes. PP1α, β and γ bound B56γ in yeast two-hybrid assays, and PP1 co-immunoprecipitated with Flag epitope-tagged B56γ and B56δ expressed in mitotic human cells. Each of these interactions was abolished by mutation of the PP1-docking-site motif (Fig. 4k and Extended Data Fig. 9b–d). Thus, the principle of a phosphatase relay, whereby PP1 recruitment activates PP2A to control mitosis, is conserved in human cells. It is notable that the NXS Polo kinase consensus sequences in fungal B56 subunits are replaced by SP and SQ motifs in metazoa (Fig. 2a), suggesting that CDK/MAP kinases modulate PP1 docking to B56δ and B56γ, whereas ATM/ATR/DNA-PK may coordinate docking to B56 α, β, ε. Furthermore, these serine/threonine residues also conform to consensus motifs for AGC kinases, including aurora B (KXXS/T), while the threonine highlighted in yellow in Fig. 2a could facilitate control by further kinases. We believe, therefore, that PP1 regulation of PP2A-B56 activity will emerge as a ubiquitous feature in diverse signalling contexts. Although bulk biochemical assays reveal core principles of phosphatase control, local controls sit at the heart of many transitions26. Thus, we propose that competition between kinases and PP1 and PP2A holoenzymes at specific locations will generate locally active, dephosphorylated phosphatases at times when assays show the activity of the bulk population is depressed.

The direct recruitment of PP1 to B55 and B56 subunits that we observed is unanticipated. Although the majority of predicted PP1-docking sites reside within regions that are predicted to be intrinsically disordered11, PP1 recruitment to the docking sites of B55Pab1, B56Par1 and CENP-E11,27 suggest that this requirement is not axiomotic. A second conundrum is that the PP1-docking motifs are inaccessible in structures of the PP2A holoenzyme complexes that have been solved so far28,29,30. PP1-docking site residues, and the equivalent residue to S378, form bonds that are crucial to the integrity of these structures, yet, contrary to predictions arising from these structures, mutation of these residues to alanine has no impact on the integrity of either PP2A-B55 or PP2A-B56 holoenzymes (Extended Data Fig. 10). Moreover, S378 is phosphorylated in purified PP2A-B56Par1 complexes and holoenzymes isolated by one-step immunoprecipitation (Extended Data Figs 5g and 10c). Our data therefore add to established discrepancies between in vivo and in vitro PP2A holoenzyme assembly24 to suggest that alternative structures will emerge from structural studies of active enzymes that have undergone the extensive array of modifications required for activity that impact upon PP2A conformation in vivo24.

Methods

Standard procedures31,32 were used to grow and maintain yeast strains (Supplementary Table 1). Appropriately supplemented EMM2 synthetic medium was used throughout with the exception of TAP tag purification, for which we used 10× YES. The generation of size-selected synchronous cultures was as described previously33. Two-hybrid assays used the matchmaker gold system (Clontech 630489). ‘Phospho-specific’ rabbit polyclonal antibodies were generated by Eurogentec. Commercial antibodies detected CPpa1 and CPpa2 (Cell Signaling, 2038), APaa1 (Abcam, ab28350) and human B56ε (Santa Cruz sc-135223). In house polyclonal antibodies detected PP1Dis2(ref. 20) and green fluorescent protein (GFP). To detect associations between PP1Dis2 and B55Pab1 and PP1Dis2 and B56Par1 the complexes were isolated from 2 × 108 cells in the buffer (50 mM HEPES (pH 7.5), 50 mM NaF, 0.4 mM Na3VO4, 40 mM Na–β-glycerol phosphate, 1 mM EDTA, 50 mM NaCl, 0.1% NP40, 1 mM PMSF). The EnzChek (Molecular Probes E12020) or histone H3 serine 10 dephosphorylation assays34 monitored PP1Dis2 activity (Extended Data Fig. 1) with the use of recombinant rabbit PP1γ (NEB P0754S), full-length NIPP1 (Abcam, ab131705), Phos-Stop (Roche 04906845001) and potato acid phosphatase (Molecular Probes E12020). Manufacturer’s instructions were followed for EnzChek assays (Molecular Probes E12020). For Histone H3 serine 10 assays, histone H3 (Millipore 14-4-11) that had been phosphorylated by established auroraArk1 kinase assays35 was used as a substrate. The dephosphorylation reaction was conducted in 20 mM HEPES, 100 mM NaCl, 1 mM EDTA, 0.1% NP40 at 30 °C for 1 h. An established PP2A-B56Par1 activity assay used GST–Rec8391–561 phosphorylated by recombinant GST–Hhp2 (S. pombe casein kinase I) as a substrate22: PP2A-B56Par1.HA was isolated from 2 × 108 cells with 12CA5 antibody (Roche) under non-denaturing conditions. The dephosphorylation reaction was maintained at 30 °C for 1 h, in 50 mM HEPES (pH 7.5), 100 mM NaCl, 0.1% NP40, 2 mM MnCl2, 2 mM dithiothreitol (DTT). 32P levels in the substrate were determined with a Phosphoimager (BioRad). Phosphatase activity was calculated as the reduction of 32P incorporation per unit B56Par1 normalized to the activity of a standard sample from an asynchronous culture. PP2A-B55Pab1 activity was monitored by detecting phosphorylation on S377 and S378 of B56Par1with the phospho-specific antibody B56-Phos (Extended Data Fig. 7a). B56Par1.HA substrate from 6 × 108 mitotically arrested cells36 was independently isolated for three individual assays (scaling up substrate production failed owing to substrate dephosphorylation and degradation during preparation). To isolate this B56Par1.HA devoid of other subunits for PP2A-B55Pab1 assays (Extended Data Fig. 7b, right), a TCA precipitation of total protein was resuspended in a denaturing IP buffer (50 mM HEPES (pH 7.5), 50 mM NaF, 0.4 mM Na3VO4, 40 mM Na–β-glycerol phosphate, 1 mM EDTA, 200 mM NaCl, 0.2% NP40, 1 mM PMSF). The dephosphorylation assay was performed at 30 °C for 1 h (100 mM Tris (pH 7.5), 150 mM NaCl, 10 mM DTT). PP2A and PP1 enzymes were purified by established sequential TAP and immuno-affinity/peptide elution methods37. For TAP tagging, Econo-Pac Chromatography Columns (Biorad 9704652) were packed with IgG Sepharose 6 FastFlow (GE Healthcare 170969-01) or Calmodulin Affinity Resin (Agilent Technologies 214303-52). The following were used for subsequent affinity isolation: anti-HA Affinity Matrix (Roche 101677100), HA peptide (Sigma, I2149), Dynabeads A (Life Technologies) pre-loaded with PP1Dis2 antibodies20 according to manufacturer’s instructions, and the SQNWHMTPPRKNK peptide for elution of PP1Dis2 from the PP1Dis2 polyclonal antibody. Coomassie (Brilliant Blue, Sigma, B2025-1EA) stained 4–12% SDS–NUPAGE gradient gels (Life Technologies, NP20327). Size-exclusion chromatography was performed on a Superdex 200 PC 3.2/30 column (VWR) using buffer B (50 mM HEPES (pH 7.5), 100 mM NaCl, 0.1% NP-40) collecting 50 µl fractions at a flow rate of 50 µl ml−1(ref. 38). All other S. pombe techniques were performed as described previously20.

For immunoprecipitation of human complexes, tetracycline-inducible HeLa cell lines were generated using a pcDNA5/FRT/TO vector (Invitrogen). Cells were grown in advanced DMEM (Invitrogen) supplemented with 2% fetal bovine serum and penicillin/streptavidin. Cells were blocked with thymidine (Sigma-Aldrich) for 24 h and released into fresh medium with 300 nM nocodazole (Sigma-Aldrich) and 1 µg ml−1 tetracycline (EMD). Prometaphase cells were collected by mitotic shake-off after 12 h. Cells were lysed for 20 min on ice in extraction buffer A (50 mM Tris-HCl (pH 8.0), 150 mM NaCl, 0.2% NP-40, 1 mM EDTA, complete inhibitor cocktail tablet (Roche), 0.2 μM microcystin (LGC-Promotech)) and lysates were cleared by centrifugation for 15 min at 12,000g and 4 °C. Protein complexes were precipitated for 1.5 h at 4 °C with anti-Flag (M2; Sigma-Aldrich) covalently coupled to Dynabeads (Invitrogen), washed in extraction buffer, and bound proteins were eluted with LDS-sample buffer (Invitrogen) before analysis by immunoblotting38. Antibodies against the Flag epitope and PP1 (E-9; Santa Cruz sc-7482) were used at 1;1,000.