LETTERS

Selective Y centromere inactivation triggers shattering in micronuclei and repair by non-homologous end joining

Peter Ly1, Levi S. Teitz2, Dong H. Kim1, Ofer Shoshani1, Helen Skaletsky3, Daniele Fachinetti1,4, David C. Page2,3 and Don W. Cleveland1,5

Chromosome missegregation into a micronucleus can cause genomic rearrangements onto one or a few chromosome(s)3. While complex and localized genomic rearrangements1,2 known as features of chromothripsis have been recapitulated from experimental chromothripsis3, but the underlying mechanisms remain systems involving dicentric bridge formation6 or exogenous DNA unresolved. Here we developed an inducible Y damage induction7, sequencing evidence from plant models1 and centromere-selective inactivation strategy by exploiting a human cells2, and genetics in yeast8, has suggested that complex CENP-A/histone H3 chimaera to directly examine the fate of and localized rearrangements can be triggered by chromosome missegregated in otherwise diploid human cells. missegregation into aberrant compartments called micronuclei. The Using this approach, we identified a temporal cascade of exact cellular mechanism(s) for how missegregated chromosomes events that are initiated following centromere inactivation become chromothriptic remains unknown, although an attractive involving chromosome missegregation, fragmentation, and hypothesis3,9,10 involves chromosome pulverization in micronuclei— re-ligation that span three consecutive cell cycles. Following an observation initially made almost half a century ago11—followed centromere inactivation, a micronucleus harbouring the Y by incorrect reassembly of fragments through DNA repair. chromosome is formed in the first cell cycle. Chromosome Attempts to directly address these underlying mechanisms shattering, producing up to 53 dispersed fragments from a have been lacking due to experimental limitations of conventional single chromosome, is triggered by premature micronuclear cell-based methods for producing micronuclei by chemically condensation prior to or during mitotic entry of the second induced prolonged mitotic arrest to generate random chromosome cycle. Lastly, canonical non-homologous end joining (NHEJ), segregation errors. This approach precludes discriminating the but not homology-dependent repair, is shown to facilitate micronucleated chromosome from normal, nuclear chromosomes in re-ligation of chromosomal fragments in the third cycle. mitosis and prevents monitoring the fate of the initially missegregated Thus, initial errors in cell division can provoke further genomic chromosome for more than one cell generation. Moreover, mitotic instability through fragmentation of micronuclear arrest can itself produce unwanted DNA damage and/or activate DNAs coupled to NHEJ-mediated reassembly in the an apoptotic degradation event12–15. We sought to bypass these subsequent interphase. limitations by inducing the missegregation of a specific chromosome- of-interest into micronuclei that can be examined over successive Chromosome segregation errors during mitosis are a well-recognized cell cycles without mitotic perturbation. To do so, we exploited cause of numerical aneuploidy and have been implicated in the a unique feature of the human Y chromosome centromere that formation of chromosomal translocations4, both hallmark features of permitted conditional, centromere-selective control for assembly of cancer genomes. Whether—and if so, how—such mitotic errors also the kinetochore—the spindle microtubule-attachment complex that contribute to the development of complex structural rearrangements is required for chromosome segregation. are poorly understood. Chromothripsis can be detected in a broad Kinetochores are assembled on the outer surface of centromeric spectrum of human cancers3,5 and is predicted to occur during a chromatin through nucleation by centromere C (CENP-C)16, single, catastrophic event that contemporaneously generates multiple which is recruited and stably attached at centromeres through its

1Ludwig Institute for Cancer Research and Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093, USA. 2Department of Biology, Massachusetts Institute of Technology and Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA. 3Howard Hughes Medical Institute and Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA. 4Present address: Institut Curie, PSL Research University, CNRS, UMR144, F-75005, Paris, France. 5Correspondence should be addressed to D.W.C. (e-mail: [email protected])

Received 18 April 2016; accepted 2 November 2016; published online 5 December 2016; DOI: 10.1038/ncb3450

68 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

a CENP-A CENP-AC–H3

Kinetochore Kinetochore Kinetochore Inactivated CC C C centromere A B A A B A

All autosomes Y chromosome All autosomes Y chromosome + X chromosome (CENP-B-deficient) + X chromosome (CENP-B-deficient)

A CENP-A B CENP-B C CENP-C A CENP-AC–H3 chimaera

bcMale human DLD-1 cells Centromeres marked Y Y X by degron-tagged X CENP-AEYFP–AID

– Dox/IAA CENP-A: CENP-C: IAA-induced degradation of endogenous CENP-AEYFP–AID Dox-induced synthesis X X

AA of rescue CENP-AC–H3 I Y Y Depleted CENP-AEYFP–AID replaced by CENP-AC–H3 + Dox/ CENP-A: CENP-C:

Figure 1 An inducible CENP-A replacement strategy enables functional and CENP-AC−H3 (red circles). Doxycycline (dox) induces transcriptional synthesis selective inactivation of the Y chromosome centromere in human cells. of the rescue CENP-AC−H3 chimaera and auxin (IAA) triggers rapid degradation (a) A schematic depicting the strategy used to functionally inactivate the of the endogenous, auxin-inducible degron (AID)-tagged CENP-A protein. CENP-B-deficient Y centromere with a CENP-A chimaera containing the (c) Immuno-FISH images of DLD-1 cells rescued with CENP-AC−H3 following carboxy-terminal tail of histone H3 (CENP-AC−H3) that cannot directly recruit 24 h dox/IAA treatment. DNA FISH was used to spatially identify the locations CENP-C. All other chromosomes assemble kinetochores through the CENP- of the X (green) and Y (red) centromeres in DAPI-stained interphase nuclei B-dependent pathway. (b) A schematic depicting the approach for inducible combined with immunofluorescence for detection of CENP-A or CENP-C. replacement of endogenous CENP-A (green circles) at all centromeres with Images are representative of two independent experiments. Scale bar, 5 µm. direct binding to the centromeric histone H3 variant CENP-A17 p53-deficient, thereby enabling chromosome missegregation and/or and the DNA sequence-specific binding protein CENP-B18. The DNA damage without eliciting a p53-dependent cell cycle arrest24,25. amino- and carboxy-terminal tails of CENP-A mediate centromeric We first stably integrated a encoding TIR1, the plant E3 ubiquitin recruitment of CENP-C (Fig. 1a) through redundant CENP-B- ligase mediating auxin-dependent target protein destruction26,27. dependent and -independent mechanisms, respectively18–21. Since This was followed by TAL effector nuclease-mediated gene editing either of these CENP-A tails is sufficient for nucleating kinetochore to disrupt one endogenous CENPA allele and to amino-terminally assembly and subsequent chromosome segregation, we previously tag the CENP-A encoded by the second allele with an enhanced proposed that the CENP-B-dependent pathway operates as a ‘back-up’ yellow fluorescent protein fused to an auxin-inducible degron mechanism to reinforce centromere function18. CENP-B specifically (CENP-A−/EYFP−AID), the latter of which permits rapid and complete binds to repetitive 17-base-pair motif sequences, termed CENP-B degradation26,27 of AID-tagged CENP-A within ∼90 min after boxes, interspersed throughout the alphoid repeats found in all addition of the plant hormone auxin (indole-3-acetic acid, IAA) human centromeres with the single exception of the Y chromosome (Supplementary Fig. 1c). Finally, we stably integrated a single centromere22,23. Recognizing this distinct feature, we reasoned copy, doxycycline (dox)-inducible gene encoding the CENP-AC−H3 that complete replacement of CENP-A at all centromeres with chimaera (or CENP-AWT (wild-type) as a control) whose basal level a chimaeric CENP-A variant in which the CENP-C-recruiting, of transcription produced CENP-AC−H3 at ∼10% of normal CENP-A carboxy-terminal tail (spanning six amino acids) is substituted with levels (Supplementary Fig. 1d–f). the corresponding region of histone H3 (hereafter referred to as Induced destruction of CENP-AEYFP−AID simultaneous with in- CENP-AC−H3, Supplementary Fig. 1a)19 would selectively inactivate duced transcription of CENP-AC−H3 following dox/IAA addition led the Y centromere while leaving its position epigenetically marked by to rapid loss of endogenous CENP-A and its replacement with CENP- chromatin incorporating CENP-AC−H3 (Fig. 1a). AC−H3 within one cell cycle28 at every centromere without loss of cen- To engineer a cell-based system for Y centromere inactivation, we tromere identity (as indicated by co-localization with anti-centromere employed a replacement strategy for removal of endogenous CENP-A antibodies—Supplementary Fig. 1e). Whereas CENP-AEYFP−AID de- by induced proteolysis and simultaneous transcriptional induction of pletion alone resulted in complete lethality, its replacement with CENP-AC−H3 (Fig. 1b and detailed in Supplementary Fig. 1b). The CENP-AC−H3 rescued broad centromere function and cell viability male human DLD-1 cell line was chosen as it was known to maintain without affecting clonogenic growth (Supplementary Fig. 1g), pro- a diploid karyotype with a low basal rate of spontaneous micronucleus liferation rate (∼25–27 h per doubling—Supplementary Fig. 1h-i) or formation (<3% of cells) and to be both mismatch repair- and cell cycle distribution (Supplementary Fig. 1j)—all of which were

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 69

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

abc Percentage of Percentage of Estimated rate of mitoses filmed mitotic errors 100 30 ∗∗∗ ∗∗∗ ∗∗ Y loss per division cells

+ 80 42.8% 70% 30% 20 13.2% 60 682 390 ∗∗∗∗ ∗∗∗∗ events events 40 NS ∗∗∗∗ 10 20 57.2% 86.8% Percentage of

Y retained with micronuclei

0 Y loss Percentage of cells 0 Normal division Lagging chromosome Y chromosome 0358 01246 Division with error Misaligned chromosome Days after dox/IAA Days after dox/IAA treatment treatment

d CENP-AWT CENP-AC–H3 H2B-mRFP – Dox/IAA + Dox/IAA – Dox/IAA + Dox/IAA Normal Misaligned Normal Lagging metaphase metaphase anaphase anaphase 100 Chromosome Y 100 Chromosome 4

75 75 e CEN-Y 24.5% 32.4% 16.7% 10.8% 50 50 CEN-4

25 25 Chromosome 4 micronuclei (%) micronuclei (%) Chromosome Y 0 0

0 5 10 15 20 25 0 5 10 15 20 25 1 2 3 4 Cells with micronuclei (%) Number of Y centromere foci

f g In vitro purified micronuclei + FISH Purify micronuclei by DAPI 50 μm Chr. Y Merge differential centrifugation Exp. 1 Exp. 2 Exp. 3

52% 44% 46% 48% 56% 54% Sucrose gradient 416 141 193 [1–1.8 M] Non-Y chromosome Nuclei Y chromosome

Figure 2 Y centromere inactivation triggers Y chromosome missegregation staining to quantify the percentage of micronucleated cells (x axis) and cross- and accumulation into micronuclei. (a) Percentage of Y chromosome- plotted against the percentage of micronuclei containing either chromosome positive signals in the primary nucleus measured by centromere FISH. Y or 4 (y axis) as measured by centromere FISH. Data represent the Lines represent the mean ± s.e.m. of n = 6 (0 d, 1,758 cells) and n=3 mean ± s.e.m. of n = 3 independent experiments per axis, and statistical independent experiments (3–8 d, 672–1,605 cells per condition). The right analyses and sample sizes for each axis are provided in Supplementary schematic depicts how Y loss is perpetuated over repeated doublings Fig. 2a,b. (e) Representative images of 5 d CENP-AC−H3-rescued cells probed within a population. (b) Percentage of micronucleated cells measured by by centromere FISH for chromosomes Y (red) and 4 (green). The percentage DAPI staining. Lines represent the mean ± s.e.m. of n = 3 independent represents the distribution of micronuclei harbouring ≥1 Y centromere foci experiments (2,045–3,153 cells per condition). Asterisks in a,b indicate (102 micronuclei pooled from 3 independent experiments). Counterstain with significance by two-tailed Student’s t-test compared with untreated control DAPI is shown in blue. Scale bar, 5 µm. (f) Representative image of DAPI- cells. ∗∗∗∗P <0.0001; ∗∗∗P < 0.001; ∗∗P < 0.01; NS, not significant. (c) 2 d stained micronuclei purified from 4 d CENP-AC−H3-rescued cells hybridized to CENP-AC−H3-rescued cells stably encoding histone H2B-mRFP were filmed Y chromosome painting probes (green) by FISH. (g) Pie charts depicting the by time-lapse microscopy for chromosome segregation errors. Data represent proportion of purified micronuclei with Y chromosome-positive FISH signals. the mean of n = 3 independent experiments. Scale bar, 5 µm. (d) Cells Numbers below charts indicate the number of micronuclei analysed per rescued with CENP-AWT or CENP-AC−H3 for 5 d were analysed by DAPI experiment. Source data for a,b,c,g are provided in Supplementary Table 1. comparable to untreated parental cells or those rescued by CENP-AWT. micronuclei within 48 h of CENP-A replacement (Fig. 2b), whereas CENP-AC−H3-rescued cells had normal mitotic duration (41 ± 11 min, elevated micronucleation was not observed in control cells or those n = 50 H2B-mRFP-labelled mitoses filmed) and were also capable rescued with CENP-AWT (Supplementary Fig. 2a). By the second of sustaining mitotic arrest when challenged with the microtubule day after CENP-AC−H3 replacement, more than half of mitoses inhibitor nocodazole (Supplementary Fig. 1j). (57 ± 5%) developed chromosome segregation errors, the large As expected, after induced degradation of CENP-AEYFP−AID majority (87 ± 5%) of which were accompanied by defects in initial and its replacement with CENP-AC−H3, the essential kinetochore- chromosome alignment (Fig. 2c). nucleating protein CENP-C was selectively lost from the Y centromere FISH probes targeting the Y centromere and the centromere of a (Fig. 1c) despite continued marking of Y centromere position control autosome (chromosome 4) were used to establish that most by CENP-AC−H3. Correspondingly, the Y chromosome underwent micronuclei (527/754 micronuclei examined, or 70 ± 10%, P =0.008) rapid population-wide loss from primary nuclei at a rate of contained the Y chromosome (Fig. 2d and Supplementary Fig. 2b), ∼30% per cell division (as measured by a Y centromere-bound while micronucleated chromosome 4 was rarely observed (<3%). fluorescent in situ hybridization (FISH) probe, Fig. 2a). Almost A ∼35-fold enrichment in CENP-AC−H3 cells with Y-containing a quarter (23 ± 1%) of CENP-AC−H3-rescued cells contained micronuclei (Supplementary Fig. 2c) was measured compared with

70 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

a Intact Y chromosome b NS NS DAPI Chr. Y CEN-Y Merge 40 P = 0.0016 ∗∗∗ ∗∗ 30 ∗∗∗ 20 10 NS NS – Dox/IAA Y chromosome 0 fragmentation (%) 012345 Days after dox/IAA treatment 110 102 104 103 93 93 Fragmented Y chromosome DAPI Chr. Y CEN-Y Merge c 0 d Dox/IAA 3 d Dox/IAA 326 spreads 317 spreads Percentage of Y chromosome+ >99% 81 ± 6% metaphase spreads Intact Y 19%

+ Dox/IAA (3 d) Fragmented Y P = 0.0037

d e 3 d Dox/IAA: fg + + 100 0 d Dox/IAA 100 Non-fragmented 60 40 spreads 75 3 d Dox/IAA 75 30 40 P < 0.0001 Fragmented 50 50 spreads 20 20 25 25 10 Percentage of Y Percentage of Y 0 0 0 0 detected per event metaphase spreads metaphase spreads Percentage of events 01234>5 01234>5 Number of fragments n = 60 012345678 Number of intact Y chromosome(s) Number of intact Y chromosome(s) Number of CEN-Y foci

hi P = 0.0423 100 + 100 nM Day: 021 30 calyculin A P = 0.026 P = 0.0299 50 Dox/IAA Async. 0 d Dox/IAA 20 PD-0332991 G1 P = 0.0002 2 d Dox/IAA nuclei (%) Condensed 0 RO-3306 G2 NS (min) 0 15304560 = Untreated 10 = Treated p-H3 (Ser10) Y chromosome

= Calyculin A (60 min) fragmentation (%) 0 Calyculin A-induced Total H3 Async. G1 arrest G2 arrest

Figure 3 Chromosomes in micronuclei are susceptible to extensive and 3 d CENP-AC−H3 rescue and FISH analysis. Data represent the mean ± catastrophic shattering in mitosis. (a) Representative FISH images of s.e.m. of n=3 independent experiments (255 non-fragmented spreads and metaphase spreads prepared from 0 d or 3 d CENP-AC−H3-rescued cells co- 62 fragmented spreads total). (f) The number of detectable Y fragments hybridized to Y chromosome paint probes (green) and Y centromere probes produced per fragmentation event was quantified and the red line indicates (red). Lower example shown contains ∼18 detectable Y fragments and the mean of 60 events. (g) Distribution of the number of Y centromere one centromere signal (red arrow). Scale bar, 10 µm. (b) Quantification of signals produced per fragmentation event (60 events scored). (h) Addition of fragmentation frequency over the indicated time intervals. Data represent calyculin A to asynchronous cells induces rapid chromosome condensation the mean ± s.e.m. of n = 3 independent experiments, and the total (top) and histone H3 phosphorylation (bottom). DAPI-stained DLD-1 cells number of metaphase spreads analysed across the three experiments are were examined for condensation and metaphase-like appearance from >135 indicated below the graph. (c) Metaphase spreads collected from 0 d nuclei per time point. (i) Calyculin A was used to induce premature or 3 d CENP-AC−H3-rescued cells were analysed for Y fragmentation as chromosome condensation (as indicated on the left) followed by spread in a,b and represent the mean of n = 3 independent experiments. Data preparation and processing for FISH as in a. Data represent the mean ± from d–g are compiled from spreads collected in c.(d) Distribution of s.e.m. of n=4 independent experiments (162–276 spreads per condition). intact Y chromosome copy number by FISH analysis. Data represent the Asterisks and P values for b,c,i indicate significance by two-tailed Student’s mean ± s.e.m. of n = 3 independent experiments (317–326 spreads t-test compared with untreated control cells or as denoted. ∗∗∗P < 0.001; pooled from 3 experiments) and P value indicates significance by two- ∗∗P < 0.01; NS, not significant. Source data for b,c,d,e,h,i are provided in way ANOVA. (e) Distribution of intact Y chromosome copy number derived Supplementary Table 1. Unprocessed original scans of blots are shown in from fragmentation-negative or -positive metaphase spreads following Supplementary Fig. 6. untreated CENP-AWT-rescued cells, with a proportion (∼49%) the Y chromosome, micronuclei were purified using differential of micronuclei containing two or three Y centromere signals centrifugation30 and analysed by FISH with a Y chromosome per micronucleus (Fig. 2e and Supplementary Fig. 2d). These painting probe (Fig. 2f). FISH analysis revealed that 52 ± 4% of micronuclei were often larger in size (R2 = 0.70, Supplementary purified micronuclei contained Y-specific sequences (Fig. 2g) without Fig. 2e), probably indicating replication of an initial micronuclear detectable contamination of primary nuclei, and the remainder chromosome29 and/or the entrapment of multiple Y chromosomes were suspected to entrap a distribution of randomly missegregated into the same micronucleus—perhaps through nondisjunction chromosomes. We conclude that selective inactivation of the Y of sister chromatids. To further confirm selective enrichment of centromere efficiently produces micronuclei that are specifically

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 71

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS enriched for the human Y chromosome without compromising other in non-micronucleated control cells regardless of cell cycle posi- major cellular characteristics (summarized in Supplementary Fig. 2f). tion (Fig. 3i). In contrast, addition of calyculin A to micronucleated As nuclear envelope disruption is a common fate for micronu- cells in interphase produced fragmented Y chromosomes prematurely clei31, a fluorescent protein fused to a nuclear localization signal (Fig. 3i), consistent with mitotic entry as the trigger for shattering (2xRFP-NLS) was used to measure compartmentalization between in- or detection of shattering that had occurred during interphase. Use duced Y chromosome micronuclei and those spontaneously generated of cell synchronization (Supplementary Fig. 4e) revealed that caly- from a low basal rate of segregation errors entrapping mostly non-Y culin A promoted fragmentation of the Y chromosome in cells in chromosomes. Both sources of micronuclei underwent disruption at G2, but not in G1 (Fig. 3i). Thus, condensation-induced fragmen- comparable frequency (∼26%, Supplementary Fig. 3a), indicative of tation was dependent on passage into or through S-phase, consis- similar nuclear membrane integrity in the Y and non-Y micronuclei. tent with the hypothesis that disruption during interphase ‘primes’ Moreover, using immunofluorescence for γ H2AX as a marker for the micronuclear chromosome(s) for shattering in mitosis2,11,31. An DNA double-strand breaks, a spectrum of micronucleus-specific DNA alternative shattering mechanism could be cytoplasmic accumula- damage was identified in ∼30% of micronuclei that ranged from a tion of active cyclin B1-Cdk134 complexes in late G2 that may pre- single focus to extensive damage (Supplementary Fig. 3b-c). maturely shuttle into disrupted micronuclei to initiate micronuclear To directly test if and when micronucleated chromosomes under- chromosome condensation and fragmentation in interphase prior to went fragmentation, a dual-coloured FISH assay was performed on mitotic entry. mitotic spreads using DNA probes spanning the entire Y chromosome Despite continued cell proliferation (Supplementary Fig. 1h), the (green) and the Y centromere (red). Y chromosome fragmentation was frequency of Y chromosome fragmentation remained constant be- defined by a spread containing multiple, scattered DNAs (visualized tween days 3–5 after centromere inactivation (Fig. 3b), suggesting by DAPI) that hybridized to Y chromosome painting probes and ≥1 possible cycles of shattering coupled with reassembly of fragments centromere signal(s) that also bound the painting probe. Although mi- into intact chromosomes. To test this hypothesis and to identify the cronuclei were present two days after CENP-A replacement (Fig. 2b), potential repair pathway(s), we assessed whether Y chromosome frag- Y chromosomes remained largely intact in mitosis (Fig. 3a). One day ments from the first mitotic cycle (day 3) would persist into the second later (day three), abundant fragmentation (24 ± 3% of Y chromosome- mitosis (day 4) (Fig. 4a) following inhibition of each of the three positive spreads, P = 0.0003) appeared (Fig. 3a,b and Supplementary mammalian DNA double-strand break repair mechanisms35: canon- Fig. 4a,b), indicating that shattering requires at least one complete cell ical non-homologous end joining (NHEJ), homologous recombina- cycle following initial micronucleus formation. tion (HR), and microhomology-mediated end joining (MMEJ). Two Following three-day centromere inactivation, copy number analy- essential components from each distinct pathway were individually sis from >300 metaphases with a detectable Y chromosome (Fig. 3c) suppressed using small interfering RNAs (siRNAs) (Fig. 4b,c). Inhi- revealed frequent Y chromosome aneuploidy (Fig. 3d), although bition of NHEJ by reductions in DNA Ligase IV (LIG4) or DNA-PKcs additional copies of intact Y chromosome(s) rarely accompanied resulted in a twofold increase in Y fragmentation frequency in the spreads with Y fragmentation (Fig. 3e). Sixty shattering events were second mitotic cycle (Fig. 4d,e), but neither interfered with cell pro- examined in detail; each generating between 3 and 57 chromosomal liferation (Fig. 4f) nor affected fragmentation in the first mitotic cycle FISH fragments large enough to be detected microscopically with an with or without centromere inactivation (Fig. 4g). Pharmacological average of 18 fragments per event (Fig. 3f). Approximately 35% of inhibition of LIG4 or DNA-PK similarly prevented reassembly of fragmented metaphases contained a single centromere focus (with up chromosomal fragments (Fig. 4h). to 53 Y chromosome fragments), whereas others harboured ≥2 foci In contrast to NHEJ, inhibition of HR or MMEJ by depletion of (Fig. 3g), indicative of either a break within the centromeric region BRCA2/RAD51 or LIG3/PARP-1, respectively, or treatment with a and/or co-fragmentation of multiple Y chromosomes. There was a RAD51 inhibitor, had no effect on fragment reassembly (Fig. 4e,h). weak correlation (R2 = 0.22) between the number of fragments gen- Given that suppression of NHEJ alone is sufficient to prevent erated and the number of centromere signals present (Supplementary fragment repair (Fig. 4d,e,h), shattered micronuclear DNAs may be Fig. 4c), consistent with the majority of events produced from one poor substrates for recognition and/or processing by components of Y chromosome. Additionally, mitoses with overt fragmentation of a HR and MMEJ. Thus, canonical NHEJ is the predominant DNA repair non-Y chromosome were rare (4/643 spreads examined). mechanism that facilitates the re-joining of micronuclei-derived chro- Micronuclei are prone to nuclear envelope disruption during in- mosome fragments, an outcome consistent with the sequence junc- terphase that terminates normal nuclear function31—a defect that can tions of chromothriptic events identified in human cancer genomes3,36, provoke delayed or stalled replication in S-phase9,11 and/or the acquisi- congenital disorders37, and experimentally derived breakpoints1,2— tion of DNA damage9,31,32 throughout interphase—suggesting that one the majority of which lack stretches of homology or microhomology. mechanism for chromosome fragmentation is the sudden compaction Prior sequencing efforts had suggested that chromothriptic- following mitotic entry of incompletely replicated or unrepaired mi- like reassembly in the primary nucleus is surprisingly efficient2, cronuclear DNAs33. To test this, the PP1/PP2A phosphatase inhibitor although a proportion of micronuclei persist throughout mitosis calyculin A was added to interphase cells containing micronuclei prior and fail to reincorporate into the nucleus9. Whether—and if so, to to shattering (2 d dox/IAA treatment—Fig. 2b and Fig. 3b) to drive what extent—micronucleus-specific DNA repair activity contributes abrupt, premature chromosome condensation accompanied by Ser10 to fragment re-ligation remains unknown. To determine whether phosphorylation of histone H3 (Fig. 3h and Supplementary Fig. 4d). fragment reassembly occurs within micronuclei, DNAs extracted from Calyculin A alone was insufficient to fragment the Y chromosome purified micronuclei (following 4 d dox/IAA treatment, Fig. 2f,g)

72 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

ab c Micronuclear chromosome MocksiControlsiLIG4siDNA-PKcssiBRCA2siRAD51 100∗ DNA ligase IV Day: 0123 4 3 d Dox/IAA DNA-PKcs MocksiControlsiLIG3siPARP1 Dox/IAA Fragmentation 245 100 DNA ligase III Inhibition of siRNA Figs 4d,e BRCA2 135 PARP-1 DNA repair 245 Inhibitor Fig. 4h 32 RAD51 GAPDH Reassembly 4 d Dox/IAA 32 = Untreated = Treated 32 GAPDH Mr (K) Mr (K) de4 d Dox/IAA + siRNA f 4 d Dox/IAA + DNA repair pathway inhibited: LIG4 siRNA 50 P = 0.0289 P = 0.0053 None 100 40 NS NS NS NS Non-homologous end joining 30 Homologous recombination 50 20 Microhomology-mediated end joining 0 0.2%

10 Cell number (%) Y chromosome

fragmentation (%) 0 Mock Control LIG4 DNA-PKcs BRCA2 RAD51 LIG3 PARP1 : siRNA siLIG4 siUBB Chr. Y/CEN-Y/DAPI siControl 96 91 91 92 90 90 69 siDNA-PKcs 4 d Dox/IAA + gh4 d Dox/IAA + inhibitor DNA-PKcs siRNA + siRNA only 3 d Dox/IAA + siRNA 50 50 P = 0.0026 40 40 P = 0.0439 30 P = 0.0028 30 NS 20 NS NS 20 10 10 Y chromosome Y chromosome fragmentation (%) 0 fragmentation (%) 0 Chr. Y/CEN-Y/DAPI Control LIG4 DNA-PKcs Control LIG4 DNA-PKcs : siRNA DMSO LIG4 DNA-PKcs RAD51 94 110 99 94 96 97 124 91121 92

Figure 4 Shattered chromosomal fragments are re-ligated by canonical non- with the indicated siRNAs and normalized to mock-transfected cells. siUBB, homologous end joining. (a) A schematic depicting hypothesis of turnover siRNA targeting ubiquitin B. Data represent the mean of biological triplicates. between fragmentation and reassembly. (b) Depletion of target protein as (g) Quantification of metaphase spreads with Y chromosome fragmentation confirmed by immunoblotting 3 d post-siRNA transfection in DLD-1 cells. with or without 3 d CENP-AC−H3 rescue following 3 d siRNA transfection. Data The asterisk indicates a nonspecific band from the anti-LIG4 antibody. Blots represent the mean ± s.e.m. of n = 3 independent experiments, and the are representative of two independent experiments, and unprocessed original number of total metaphase spreads analysed are indicated. (h) Quantification scans are shown in Supplementary Fig. 6. (c) Experimental schematic used of Y chromosome fragmentation from 4 d CENP-AC−H3-rescued cells treated for the indicated panels. A detailed timeline for each experiment is provided with chemical DNA repair inhibitors as indicated in c. Data represent in Supplementary Fig. 4f. (d) Representative metaphase FISH images of the mean ± s.e.m. of n = 3 independent experiments (LIG4, RAD51) or Y chromosome fragmentation events derived from 4 d CENP-AC−H3-rescued n = 4 independent experiments (DMSO, DNA-PK), and the numbers of cells 3 d post-transfection with the indicated siRNAs. Scale bars, 10 µm. total metaphase spreads analysed are indicated. Significance for e,g,h are (e) Quantification of d. Data represent the mean ± s.e.m. of n=3 independent determined by two-tailed Student’s t-test compared with control siRNAs or experiments, and the number of total metaphase spreads analysed are shown DMSO conditions or as denoted. NS, not significant. Source data for e,f,h are below the graph. (f) DLD-1 cell numbers were counted 3 d post-transfection provided in Supplementary Table 1. were subjected to paired-end sequencing to search for and quantify estimated as a polynomial function of chromosome concentration potential intra-chromosomal re-ligation events. Genomic DNAs from within a given sample (a second-order rate reaction, Supplementary parental cells were sequenced in parallel as a reference, and paired Fig. 5c). For the Y chromosome, discordant pairs were detected reads of equivalent size and quality were generated from each source of slightly below an expected frequency—a reduction that may reflect DNA (Supplementary Fig. 5a,b). Consistent with prior measurements the complex sequence features of the human Y chromosome39. As by FISH (Fig. 2g), purified micronuclear fractions were selectively sequencing of micronuclear fractions excludes chromosomes that enriched for the Y chromosome, as indicated by an average ∼27-fold were subjected to repair after nuclear incorporation, these data suggest enrichment in Y-mapped sequencing reads (Fig. 5a). Due to co- that NHEJ-dependent re-ligation spatially occurs primarily in the sedimentation with micronuclei, these fractions were also enriched main nucleus following fragment reincorporation. Thus, consistent (∼300-fold) for mitochondrial DNA sequences that were excluded with terminated nuclear function within disrupted micronuclei31, from further analyses. repair activity in micronuclei either does not occur or occurs at an Discordant sequencing pairs—a readout for structural rear- efficiency too low to be detected by sequencing. rangements38—were quantified for each sample and defined as The approach reported here can in principle be used to inactivate reads whose paired-ends mapped >100 kilobases apart on the same any CENP-B-deficient centromere, in particular rare chromosomes chromosome. Consistent across three independent experimental containing neocentromeres. Applying this strategy for the Y chromo- samples for every micronuclear chromosome, discordant pairs were some, we have determined the fate of missegregated chromosomes not detected significantly above a level expected from random over subsequent cell generations and have reconstructed the sequence re-ligation events that arise artificially during next-generation DNA of major events underlying chromothripsis involving chromosomal library construction (Fig. 5b). The number of discordant background micronucleation, fragmentation, and reassembly. Our results support events (which accounts for ∼0.05% of total sequencing pairs) can be a multi-cell cycle mechanism for chromothripsis (Fig. 5c) in which

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 73

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

Enrichment in total sequencing pairs ab50 Discordant sequencing pairs Genomic DNA (n = 3) Micronuclear DNA (n = 3) 40 Genomic DNA (n = 3) Exp. 3 6,000 4,000 Micronuclear DNA (n = 3) 2 2 30 R = 0.99 R = 0.98 Exp. 2 20 Exp. 1 3,000 2,000 10 Exp. 3 Observed Observed Exp. 2 Fold change over Exp. 1 0 0 0 average of genomic DNA 1 2 3 4 5 6 7 8 9 X 10 11 12 13 14 15 16 17 18 19 20 21 22 Y 0 3,000 6,000 0 2,000 4,000 Chromosome Expected Expected

c CENP-AC–H3 loading G1 S G2 Mitosis at centromeres Chromosome alignment Cell cycle 1 Normal failure Y chromosome

Inactivated Y centromere

Inefficient DNA repair Fragments sequestered in a in micronuclei non-disassembled micronucleus G2 S G1

Y chromosome loss Cell cycle 2 DNA shattering in mitosis Defective replication Micronucleus Y chromosome in Fragments and/or unrepaired damage disruption intact micronucleus distributed into one or both Micronucleus disassembles and daughter nuclei fragments spill into mitotic cytoplasm

G1 Fragments re-ligated by canonical NHEJ Sustained Reincorporated Y centromere Cell cycle 3 Derivative fragments activate inactivation Y chromosome DNA damage repair LIG4- dependent

Figure 5 Repair by non-homologous end joining does not occur efficiently number of discordant pairs produced from random ligation events within micronuclei. (a) Enrichment in the number of paired-end sequencing during library construction that were extrapolated from the graphs in reads mapping to each chromosome. Each data point represents an Supplementary Fig. 5c. Each dot represents a single chromosome from independent DNA sample, and micronuclear DNAs were obtained from n = 3 independent samples derived from the indicated DNA source, the purified fractions shown in Fig. 2f,g. The total numbers of and the red dots represent the Y chromosome. (c) A model depicting reads are provided in Statistics Source Data (Supplementary Table 1). how micronucleation, shattering, and NHEJ-mediated repair facilitates (b) Discordant pairs (with read-ends mapping >100 kilobases apart on chromothripsis on initially missegregated chromosomes within three the same chromosome) were quantified and plotted against an expected consecutive cell cycles.

a missegregated chromosome entrapped into a micronucleus first ACKNOWLEDGEMENTS undergoes shattering induced through mitotic entry-driven prema- We thank K. Jepsen and the UC San Diego IGM Genomics Center (MCC P30 CA023100) for DNA library preparation and sequencing, A. Shiau, S. Dowdy, ture micronuclear chromosome condensation. In the next cell cycle, E. Hatch, M. Hetzer, X. Wu and T. Pyntikova (Whitehead Institute, USA) for the resulting fragments are incorporated into one or both newly providing reagents, D. Jenkins, I. Goyal and Y. Sun for technical assistance, and formed daughter nuclei—perhaps through one or more undetermined the UC San Diego School of Medicine Microscopy Core (NINDS P30 NS047101) for shared use of equipment. This work was funded by a grant from the NIH DNA tethering mechanism(s)—and efficiently re-ligated in random (GM029513) to D.W.C., who receives salary support from the Ludwig Institute for order by LIG4 in a manner analogous to translocation formation in Cancer Research. D.C.P. is supported by the Howard Hughes Medical Institute and human cells40. We propose that canonical NHEJ, which joins DNA NIH (HG007852). P.L.was supported by a Cancer Cell Biology Training Grant from the NCI (5T32CA067754-18) and a Postdoctoral Fellowship from the Hope Funds breaks independent of , produces structural re- for Cancer Research (HFCR-14-06-06). arrangements on chromosomes initially missegregated from two cell cycles earlier.  AUTHOR CONTRIBUTIONS P.L. and D.W.C. conceived the project, designed the experiments, and wrote the manuscript. P.L. conducted the experiments and analysed the data. D.F. constructed METHODS the parental AID-tagged CENP-A cell line and provided key experimental input. Methods, including statements of data availability and any associated P.L. and D.H.K. performed purification of micronuclei. O.S. assisted with FISH accession codes and references, are available in the online version of experiments. L.S.T., H.S. and D.C.P. analysed the sequencing data. All authors contributed comments on the final manuscript. this paper. COMPETING FINANCIAL INTERESTS Note: Supplementary Information is available in the online version of the paper The authors declare no competing financial interests.

74 NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ LETTERS

Published online at http://dx.doi.org/10.1038/ncb3450 22. Masumoto, H., Masukata, H., Muro, Y., Nozaki, N. & Okazaki, T. A human Reprints and permissions information is available online at www.nature.com/reprints centromere antigen (CENP-B) interacts with a short specific sequence in alphoid DNA, a human centromeric satellite. J. Cell Biol. 109, 1963–1973 (1989). 1. Tan, E. H. et al. Catastrophic chromosomal restructuring during genome elimination 23. Earnshaw, W. C. et al. Molecular cloning of cDNA for CENP-B, the major human in plants. eLife 4, e06516 (2015). centromere autoantigen. J. Cell Biol. 104, 817–829 (1987). 2. Zhang, C.-Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 24. Thompson, S. L. & Compton, D. A. Proliferation of aneuploid human cells is limited 179–184 (2015). by a p53-dependent mechanism. J. Cell Biol. 188, 369–381 (2010). 3. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single 25. Hinchcliffe, E. H. et al. Chromosome missegregation during anaphase triggers p53 catastrophic event during cancer development. Cell 144, 27–40 (2011). cell cycle arrest through histone H3.3 Ser31 phosphorylation. Nat. Cell Biol. 18, 4. Janssen, A. et al. Chromosome segregation errors as a cause of DNA damage and 668–675 (2016). structural chromosome aberrations. Science 333, 1895–1898 (2011). 26. Holland, A. J., Fachinetti, D., Han, J. S. & Cleveland, D. W. Inducible, reversible 5. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic system for the rapid and complete degradation of in mammalian cells. Proc. DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012). Natl Acad. Sci. USA 109, E3350–E3357 (2012). 6. Maciejowski, J., Li, Y., Bosco, N., Campbell, P. J. & de Lange, T. Chromothripsis and 27. Nishimura, K., Fukagawa, T., Takisawa, H., Kakimoto, T. & Kanemaki, M. An auxin- kataegis induced by telomere crisis. Cell 163, 1641–1654 (2015). based degron system for the rapid depletion of proteins in nonplant cells. Nat. 7. Mardin, B. R. et al. A cell-based model system links chromothripsis with hyperploidy. Methods 6, 917–922 (2009). Mol. Syst. Biol. 11, 828–828 (2015). 28. Jansen, L. E. T., Black, B. E., Foltz, D. R. & Cleveland, D. W. Propagation 8. Sabatinos, S. A., Ranatunga, N. S., Yuan, J.-P., Green, M. D. & Forsburg, S. L. of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176, Replication stress in early S phase generates apparent micronuclei and chromosome 795–805 (2007). rearrangement in fission yeast. Mol. Biol. Cell 26, 3439–3450 (2015). 29. Leach, N. T. & Jackson-Cook, C. Micronuclei with multiple copies of the 9. Crasta, K. et al. DNA breaks and chromosome pulverization from errors in mitosis. X chromosome: do chromosomes replicate in micronuclei? Mutat. Res. 554, Nature 482, 53–58 (2012). 89–94 (2004). 10. Holland, A. J. & Cleveland, D. W. Chromoanagenesis and cancer: mechanisms and 30. Shimizu, N., Kanda, T. & Wahl, G. M. Selective capture of acentric fragments by consequences of localized, complex chromosomal rearrangements. Nat. Med. 18, micronuclei provides a rapid method for purifying extrachromosomally amplified 1630–1638 (2012). DNA. Nat. Genet. 12, 65–71 (1996). 11. Kato, H. & Sandberg, A. A. Chromosome pulverization in human cells with 31. Hatch, E. M., Fischer, A. H., Deerinck, T. J. & Hetzer, M. W. Catastrophic nuclear micronuclei. J. Natl Cancer Inst. 40, 165–179 (1968). envelope collapse in cancer cell micronuclei. Cell 154, 47–60 (2013). 12. Orth, J. D., Loewer, A., Lahav, G. & Mitchison, T. J. Prolonged mitotic arrest triggers 32. Terradas, M., Martín, M., Tusell, L. & Genescà, A. DNA lesions sequestered partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol. in micronuclei induce a local defective-damage response. DNA Rep. 8, Biol. Cell 23, 567–576 (2012). 1225–1234 (2009). 13. Ganem, N. J. & Pellman, D. Linking abnormal mitosis to the acquisition of DNA 33. Johnson, R. T. & Rao, P. N. Mammalian cell fusion: induction of premature damage. J. Cell Biol. 199, 871–881 (2012). chromosome condensation in interphase nuclei. Nature 226, 717–722 (1970). 14. Dalton, W. B. et al. Human cancer cells commonly acquire DNA damage during 34. Gavet, O., Gavet, O. & Pines, J. Activation of cyclin B1-Cdk1 synchronizes events in mitotic arrest. Cancer Res. 67, 11487–11492 (2007). the nucleus and the cytoplasm at mitosis. J. Cell Biol. 189, 247–259 (2010). 15. Hayashi, M. T., Cesare, A. J., Fitzpatrick, J. A. J., Lazzerini-Denchi, E. & 35. Ceccaldi, R., Rondinelli, B. & D’Andrea, A. D. Repair pathway choices Karlseder, J. A telomere-dependent DNA damage checkpoint induced by prolonged and consequences at the double-strand break. Trends Cell Biol. 26, mitotic arrest. Nat. Struct. Mol. Biol. 19, 387–394 (2012). 52–64 (2016). 16. Klare, K. et al. CENP-C is a blueprint for constitutive centromere-associated network 36. Malhotra, A. et al. Breakpoint profiling of 64 cancer genomes reveals numerous assembly within human kinetochores. J. Cell Biol. 210, 11–22 (2015). complex rearrangements spawned by homology-independent mechanisms. Genome 17. Carroll, C. W., Milks, K. J. & Straight, A. F. Dual recognition of CENP-A nucleosomes Res. 23, 762–776 (2013). is required for centromere assembly. J. Cell Biol. 189, 1143–1155 (2010). 37. Kloosterman, W. P. et al. Constitutional chromothripsis rearrangements involve 18. Fachinetti, D. et al. DNA sequence-specific binding of CENP-B enhances the fidelity clustered double-stranded DNA breaks and nonhomologous repair mechanisms. Cell of human centromere function. Dev. Cell 33, 314–327 (2015). Rep. 1, 648–655 (2012). 19. Fachinetti, D. et al. A two-step mechanism for epigenetic specification of centromere 38. Campbell, P. J. et al. Identification of somatically acquired rearrangements in cancer identity and function. Nat. Cell Biol. 15, 1–13 (2013). using genome-wide massively parallel paired-end sequencing. Nat. Genet. 40, 20. Logsdon, G. A. et al. Both tails and the centromere targeting domain of CENP-A are 722–729 (2008). required for centromere establishment. J. Cell Biol. 208, 521–531 (2015). 39. Skaletsky, H. et al. The male-specific region of the human Y chromosome is a mosaic 21. Guse, A., Carroll, C. W., Moree, B., Fuller, C. J. & Straight, A. F. In vitro centromere of discrete sequence classes. Nature 423, 825–837 (2003). and kinetochore assembly on defined chromatin templates. Nature 477, 40. Ghezraoui, H. et al. Chromosomal translocations in human cells are generated by 354–358 (2011). canonical nonhomologous end-joining. Mol. Cell 55, 829–842 (2014).

NATURE CELL BIOLOGY VOLUME 19 | NUMBER 1 | JANUARY 2017 75

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ METHODS DOI: 10.1038/ncb3450

METHODS chromosome fragments; and at least one acentric Y fragment must be generated. Cell culture, generation of stable cell lines, and reagents. T-REx Flp-In DLD-1 cells For calyculin A experiments, only ‘metaphase-like’ spreads that yielded distinct and (provided by S. Taylor, University of Manchester, UK) were cultured in Dulbecco’s normal appearing single (G1) or double chromatids (G2) were scored. modified Eagle’s medium (DMEM, Thermo Fisher) supplemented with 10% tetracycline-free fetal bovine serum (Omega Scientific) and 100 U ml−1 penicillin– Immunofluorescence and immuno-FISH. For indirect immunofluorescence, cells ◦ streptomycin. Cells were maintained at 37 C under 5% CO2 and atmospheric plated onto poly-L-lysine-coated coverslips were fixed in 4% formaldehyde for oxygen. CENPA alleles were genetically modified in TIR1-expressing T-REx Flp-In 10 min. Cells were pre-extracted with 0.3% Triton X-100 in PBS for 5 min and incu- DLD-1 cells26 by co-transfection with pcDNA3.1 plasmids (Invitrogen) encoding bated in Triton Block (0.2 M glycine, 2.5% FBS, 0.1% Triton X-100, PBS). The follow- TAL effector nucleases, as previously described18, and an EYFP-AID donor construct ing primary antibodies were used at 1:1,000 dilution (unless noted) in Triton Block targeting the translation start codon of CENPA. Single EYFP+ cells were isolated by and washed with 0.1% Triton X-100 in PBS: anti-CENP-A (Abcam, ab13939), anti- fluorescence-activated cell sorting (Sony SH800) and screened by immunoblotting CENP-C (MBL, PD030), 1:400 anti-centromere antibodies (Antibodies Incorpo- and PCR for CENP-A−/EYFP−AID clones. CENP-AWT and CENP-AC−H3 rescue cDNAs rated, 15-235-0001), anti-phospho H2AX (S139) clone JBW301 (EMD Millipore, were cloned into pcDNA5/FRT/TO plasmids and co-transfected with pOG44 into 05-636), and 1:200 anti-Lamin B1 (Proteintech Group, 12987-1-ap). Immunofluo- TIR1-DLD-1 CENP-A−/EYFP−AID cells using X-tremeGENE 9 (Roche). Cells that rescent images were acquired on a DeltaVision Core system at ×40–60 magnification underwent stable Flp recombinase-mediated transgene integration at the FRT locus (30 × 0.2 µm z-sections) and deconvolved maximum intensity projections were were selected with 100 µg ml−1 hygromycin (Thermo Fisher), and Y chromosome- generated using the softWoRx program. ImageJ was used to quantify fluorescent positive clones were confirmed by FISH. intensity. For immunofluorescence combined with DNA FISH (immuno-FISH), the To generate stable cell lines expressing fluorescent reporters of interest, H2B- immunofluorescence procedure was performed first followed by methanol/acetic mRFP and mCherry-NLS-TagRFP (annotated as 2xRFP-NLS, a gift from E. Hatch acid (3:1) fixation and the described FISH procedure. and M. Hetzer, Salk Institute, USA) open reading frames were cloned into pBABE retroviral vectors and packaged in 293GP cells by co-transfection with pVSV-G Immunoblotting. Whole-cell extracts were collected in SDS sample buffer and using X-tremeGENE 9. Viral supernatants after 48- or 72-h transfection were boiled for 10 min. Samples were resolved by SDS–PAGE, transferred to PVDF, filtered (0.45 µm) and target cells were infected in the presence of 5 µg ml−1 and blocked with 5% milk in PBST (PBS, 0.1% Tween-20). The following primary Polybrene (Santa Cruz) for ∼16 h. Fluorescent cells were isolated by fluorescence- antibodies were used at 1:1,000 dilution (unless noted) in PBST: anti-CENP-A activated cell sorting (Sony SH800). (Cell Signaling, 2186), anti-phospho histone H2AX (Ser139) clone JBW301 (EMD Doxycycline and the auxin plant hormone indole-3-acetic acid (IAA) purchased Millipore, 05-636), anti-phospho histone H3 (Ser10) (Cell Signaling, 9706), 1:4,000 from Sigma were dissolved in cell culture-grade water and used at 1 µg ml−1 and anti-histone H3 (Sigma H0164), anti-LIG4 (GeneTex, GTX100100), anti-DNA-PKcs 500 µM, respectively. For cell cycle arrest experiments, 100 ng ml−1 nocodazole (Bethyl, A300-516A), anti-LIG3 (Bethyl, A301-637A), anti-PARP (BD Pharmingen, (Sigma) was used for mitotic arrest, 1 µM of the CDK4/6 inhibitor PD-0332991 556362, provided by X. Wu, The Scripps Research Institute, USA), anti-BRCA2 (provided by S. Dowdy, UC San Diego, USA) was used for G1 arrest, and 10 µM (Bethyl, A303-434A), anti-RAD51 (Abgent, AM8421b), and 1:2,000 anti-GAPDH of the CDK1 inhibitor RO-3306 (Sigma) was used for G2 arrest, all of which (Cell Signaling, 14C10). Blots were probed with 1:4,000 dilutions of HRP-conjugated were dissolved in dimethylsulfoxide (DMSO). The following DNA damage repair secondary antibodies (GE Healthcare) and exposed to film. All unprocessed film inhibitors were dissolved in DMSO and used at the indicated concentrations: scans with the appropriate size markers are provided in Supplementary Fig. 6. 250 µM SCR7 (LIG4 inhibitor), 25 µM RI-1 (RAD51 inhibitor, both provided by A. Shiau, Ludwig Institute for Cancer Research, USA), and 10 µM NU7026 (DNA-PK siRNA transfection. The following SMARTpool ON-TARGETplus siRNAs were inhibitor, Abcam). purchased from GE Dharmacon and used: LIG4 (L-004254-00-0005), DNA-PK All cell lines were tested for mycoplasma and confirmed free of contamination. (L-005030-00-0005), BRCA2 (L-003462-00-0005), RAD51 (L-003530-00-0005), The cell lines used in this study were not authenticated and are not found in the LIG3 (L-009227-00-0005) and PARP1 (L-006656-03-0005). Transfections were database of commonly misidentified cell lines that is maintained by ICLAC and conducted using Lipofectamine RNAiMAX (Thermo Fisher) at a final siRNA NCBI BioSample. concentration of 20 nM. Non-targeting siRNAs (D-001810-04-05) were used for experimental controls, and siRNAs targeting ubiquitin B (UBB, L-013382-00-0005) Cell growth and clonogenic assays. For cell doubling time measurements, cells were were used to measure transfection efficiency by cell lethality (>99% cell death plated into six-well dishes in triplicate and counted at three-day intervals. For cell following 48 h transfection). cycle analysis, ethanol-fixed cells were stained with 10 µg ml−1 propidium iodide and 50 µg ml−1 RNase A and analysed for DNA content by flow cytometry on a BD LSR Live-cell imaging. DLD-1 cells expressing retrovirus-integrated H2B-mRFP were

II instrument (BD Biosciences). For clonogenic growth assays, 100 cells were plated plated in chambered slides (Ibidi) and switched to CO2-independent medium into six-well dishes in triplicate for two weeks. Methanol-fixed colonies were stained (Thermo Fisher) supplemented with 10% fetal bovine serum prior to filming by with a 0.5% crystal violet, 25% methanol solution and manually quantified. time-lapse microscopy. Images were acquired on a DeltaVision Core system in a controlled 37 ◦C environment at 4-min intervals for 12 h using ×40 magnification Mitotic spread preparation and DNA fluorescent in situ hybridization (FISH). For (10 × 4 µm z-sections) and low-powered exposures. Maximum intensity projections interphase FISH, cells plated in chambered slides were fixed in cold methanol/acetic were generated using the softWoRx program and videos were analysed in ImageJ. acid (3:1) for 15 min and dehydrated with 80% ethanol. For metaphase spreads, Mitotic timing is defined as the duration from nuclear envelope breakdown to cells were arrested for 4 h with 100 ng ml−1 colcemid (KaryoMAX, Thermo Fisher), anaphase onset. harvested by trypsinization, and incubated at 37 ◦C in hypotonic 75 mM KCl solution for 6 min. Cell pellets were then fixed with cold methanol/acetic acid (3:1) Micronuclei purification and sequencing. The procedure described in ref. 30 was and dropped onto slides. For calyculin A-induced spreads, trypsinized cell pellets closely followed for micronuclei purification with the exception of hydroxyurea were resuspended in 1 ml medium containing 100 nM calyculin A (Calbiochem) and treatment. Briefly, ∼200 million cells were collected, resuspended, and incubated incubated at 37 ◦C. After 1 h, 9 ml cold PBS was added, centrifuged, and cells were in DMEM containing 10 µg ml−1 cytochalasin B (Sigma) for 30 min at 37 ◦C, processed as described for metaphase spreads. pelleted, and gently Dounce homogenized in cold lysis buffer (10 mM Tris-HCl,

For DNA FISH, centromere enumeration and/or whole chromosome painting 2 mM magnesium acetate, 3 mM CaCl2, 0.32 M sucrose, 0.1 mM EDTA, 1 mM probes (MetaSystems) combined at equal ratio were applied to slides, sealed with dithiothreitol, 0.1% NP-40, 0.15 mM spermine, 0.75 mM spermidine, 10 µg ml−1 a coverslip, co-denatured at 75 ◦C for 2 min, and hybridized overnight at 37 ◦C in cytochalasin B, pH 8.5, 4 ◦C) with ten slow strokes of a loose-fitting pestle. Release a humidified chamber. Slides were subsequently washed with 0.4× SSC at 72 ◦C of nuclei was confirmed by DAPI staining and microscopy. The homogenate was for 2 min and rinsed in 2× SSC, 0.05% Tween-20 at room temperature for 30 s. centrifuged (Thermo Scientific Sorvall Legend XTR) through a step gradient of 1.06, Slides were then rinsed in water, counterstained with DAPI, and mounted in anti- 1.4 and 1.8 M sucrose buffer (10 mM Tris-HCl, 5 mM magnesium acetate, 0.1 mM fade solution. FISH images were acquired on a DeltaVision Core system (Applied EDTA, 1 mM dithiothreitol, 0.3% BSA, 0.15 mM spermine, 0.75 mM spermidine, Precision) at ×60 magnification (5 × 1 µm z-sections) and maximum intensity pH 8.0, 4 ◦C). The 1.8 M sucrose fraction was pelleted using ultracentrifugation projections were generated using the softWoRx program. (Beckman Coulter Optima L-80 XP), resuspended in 0.8 M sucrose buffer, and separated through a linear gradient of 1.0 to 1.8 M sucrose buffer using Quantification of fragmentation FISH. After Y chromosome paint and centromere centrifugation (Thermo Scientific Sorvall Legend XTR). Fractions (0.25–0.5 ml) FISH on metaphase spreads, Y chromosome-positive spreads were manually scored collected from the top of the gradient were examined for purity by DAPI staining. for fragmentation on the basis of the following criteria: Y chromosome paint signal Fractions containing pure micronuclei free of primary nuclei contamination were must be DAPI-positive; Y centromere signal must be DAPI-positive and overlap combined and diluted in PBS. Five per cent of the final fraction was fixed in with paint signal; each fragmentation event must generate a minimum of three Y methanol/acetic acid (3:1) and spotted onto a glass slide for FISH analysis. Genomic

NATURE CELL BIOLOGY

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ DOI: 10.1038/ncb3450 METHODS

(from untreated parental DLD-1 CENP-AC−H3) and micronuclear DNAs were rearrangements. Discordant pairs are present in sequencing data sets as a result extracted using a Quick-gDNA MiniPrep column (Zymo Research). Micronuclear of random ligation of short DNA fragments during library preparation. As this fractions typically recovered ∼40 ng of total DNA per experiment. formation is random, discordant pairs can be viewed as a second-order reaction For library preparation, DNA samples were fragmented using Adaptive Focused in which two fragments of DNA from the same chromosome randomly collide Acoustics (E220 Focused Ultrasonicator, Covaris) to produce a target average and ligate. Therefore, the number of such pairs should be proportional to the fragment size of 1,000 bp. Fragmented DNA was purified using the Agencourt square of the fraction of total reads from that chromosome—a prediction that is AMPure XP beads (Beckman Coulter) and sequencing libraries were generating supported by the genomic and micronuclear DNA sequencing data sets shown using the KAPA Hyper Prep Kit (KAPA Biosystems) following the manufacturer’s in Supplementary Fig. 5c. The predicted fraction of discordant pairs for each instructions using nine cycles of amplification. Library quality was assessed using the chromosome was calculated as f 2 ×n×18.34, where f is the fraction of total reads High Sensitivity D1000 kit on a 2200 TapeStation instrument (Agilent Technologies) from the chromosome, n is the total number of discordant pairs in the data set, and and size-selected on a PippinHT (Sage Sciences) instrument with a 1.5% agarose 18.34 is a normalization factor equal to 1 divided by the sum of the squares of the gel for 600–1,500 bp fragments. Paired-end sequencing was performed using an fraction of total reads from each chromosome. Illumina HiSeq 2500 instrument, generating 100 bp × 2 reads to a target depth of approximately 60 million paired reads per sample. Statistical analysis and reproducibility. No statistical method was used to predetermine sample size and experiments were not randomized. Investigators Discordant sequencing analysis. Sequencing read quality for all samples were were not blinded to group allocation during experiments or outcome assessment. confirmed with FastQC (http://www.bioinformatics.bbsrc.ac.uk/projects/fastqc/), GraphPad Prism 5.0 software was used to calculate statistical significance as and adapter sequences were trimmed on both ends using cutadapt41. Reads were specified in the figure legends. Graphs represent mean ± standard error (unless mapped to the hg38 human reference genome using Bowtie242 with maximum noted) and a P value of <0.05 derived from at least three independent exper- valid fragment length set to 2,000 bp. Alignment files were sorted and merged iments was considered to be statistically significant. Figures with representative using SAMtools43 to generate individual files for each sample. Duplicate reads were images were repeated independently at least twice (except for Fig. 3h, which removed using Picard’s MarkDuplicates (http://broadinstitute.github.io/picard). was performed once to identify optimal treatment time for downstream exper- DNA fragment sizes were recalculated using 1,000 mapped read pairs per iments). Data points for each quantitative experiment can be found in the chromosome, each with mapping quality ≥35 and no clipped bases. Fragment size Statistics Source Data (Supplementary Table 1). Unprocessed film scans from all was defined as the distance between the first and last bases on the reference genome immunoblotting experiments are shown in Supplementary Fig. 6 with the relevant to which either read in the pair mapped. As extreme fragment size outliers can size markers indicated. skew these calculations, and are probably the result of chimaeric fragments or true rearrangements rather than representing extremely large fragments, the top and Data availability. DNA sequencing data reported in this study have been deposited bottom 2% of fragment sizes were removed before calculating the mean and variance. in NCBI Sequence Read Archive with the primary accession code SRP074439, Read pairs with mapping quality <35 were filtered out for the discordant pair and source data for Fig. 5a,b and Supplementary Fig. 5a,b have been provided in analysis. Repeats such as LINEs and SINEs were not masked, as differences in the Supplementary Table 1. All other data that support the findings of this study are locations of such repeats are unlikely given that both genomic and micronuclear available from the corresponding author on request. DNA samples were derived from the same cell line. Discordant read pairs caused by differences between the reference genome and the genome of the cell line should 41. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing therefore be present at equal proportions in both samples. Discordant pairs were reads. EMBnet. J. 17, 10–12 (2011). defined as paired sequencing reads in which ends mapped at least 100,000 bp apart 42. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. on the same chromosome. Methods 9, 357–359 (2012). To measure whether discordant pairs were enriched in the micronuclear 43. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, samples, we considered how discordant read pairs arise in the absence of de novo 2078–2079 (2009).

NATURE CELL BIOLOGY

© ƐƎƏƖɥMacmillan Publishers LimitedƦɥ/ 13ɥ.$ɥ/1(-%#1ɥ 341#. All rights reservedƥ Correction notice Nature Cell Biology http://dx.doi.org/10.1038/ncb3450 (2016).

Selective Y centromere inactivation triggers chromosome shattering in micronuclei and repair by non-homologous end joining

Peter Ly, Levi S. Teitz, Dong H. Kim, Ofer Shoshani, Helen Skaletsky, Daniele Fachinetti, David C. Page and Don W. Cleveland

The original version of this file contained a loss of resolution in the Supplementary Figures and a loss of superscript text formatting. These errors were introduced during the production of the file and were corrected on 22 December 2016.

NATURE CELL BIOLOGY SUPPLEMENTARY INFORMATION

DOI: 10.1038Supplementary/ncb3450 Figure 1 In the format provided by the authors and unedited.

a Human Centromere Protein A (CENP-A): e No rescue CENP-AWT CENP-AC-H3 1 44 75 114 135 140 - +IAA - +IAA - +IAA amino tail CATD carboxy tail histone fold domain

CENP-A WT -DVQLARRIRGLEEGLG CENP-A CENP-A C-H3 -DVQLARRIRGERA--- 125 135 140

b T-REx Flp-In DLD-1 cells -Dox ACA

Retroviral TIR1 Puromycin selection Merge Co-transfect with CENP-A TALENs w/ DAPI + EYFP-AID targeting vector Single-cell sorting Isolate CENP-A–/EYFP-AID clone CENP-A

Integrate doxycycline-inducible CENP-AWT or C-H3 rescue into Flp-In locus Hygromycin selection +Dox ACA DLD-1 CENP-A–/EYFP-AID with doxycycline-inducible CENP-AWT or C-H3 Merge c -IAA +IAA w/ DAPI

No rescue CENP-AWT CENP-AC-H3

Histone H2BmRFP / CENP-AEYFP-AID

d WT C-H3 No rescue CENP-A CENP-A +Dox/IAA +Dox/IAA +Dox/IAA Dox: - - + + - - + + - - + + IAA: - + - + - + - + - + - + 80- Endogenous f 60- CENP-AEYFP-AID Input: 1x 1x 1x 1x 2x 4x 8x 50- Dox: + + - - - - - 40- IB: α-CENP-A 30- IAA: + - - + + + + 25- 20- CENP-A 20- Rescue 15- 15- CENP-A MW (kDa) - 40- 40 GAPDH - 30- IB: α-GAPDH 30 1 2 3 4 5 6 7 8 9 10 11 12

-Dox/IAA +Dox/IAA g h i ) 10 s 30 -Dox/IAA h r

( ) c 100 NS NS 8 +Dox/IAA e % ( m i

doub li ng s 20 h 75 6 t on i o w 50 t 4

a 10 g r Cl onogen i 25 2 ** oub li ng t opu l D

0 P 0 0 WT 3 0 2 4 6 8 10 - + e A C-H A Days after dox/IAA treatment No rescu CENP- CENP- Dox/IAA

j -Dox/IAA +Dox/IAA -Dox/IAA +Dox/IAA +nocodazole +nocodazole Events

DNA content

WWW.NATURE.COM/NATURECELLBIOLOGY 1 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION Supplementary Figure 2

Supplementary Figure 1 Construction of human DLD-1 cells with auxin- antibodies. Scale bar, 5 μm. (f) Dox-inducible CENP-A is expressed at b a -Dox/IAA +Dox/IAA p=0.0013 degradable CENP-AAID and a doxycycline-inducible CENP-AC-H3 rescue that low basal levels without supplemented doxycycline, allowing for the p=0.0016 p=0.016 is capable of maintaining centromere identity and function. (a) Amino acid simultaneous addition of dox and IAA without epigenetic loss of centromere ll s

C-H3 )

100 n

sequence of wild-type CENP-A (WT) and the carboxy-terminal tail chimera identity. (g) CENP-A -rescued cells are capable of sustaining long- i e ei p=0.0079 l 40 p=0.0004 % c o f ( e f (CH-3) swapped with the corresponding tail of histone H3. Schematic not term clonal growth and viability using a 2-week colony formation assay.

o 20 75 e i Chromosome 4 l o m drawn to scale. CATD; centromere targeting domain. (b) Schematic for Data were normalized to untreated cells and represent the mean ± SEM onu c r c o s i the construction of DLD-1 cell lines used in all experiments. (c) Unfixed of n = 3 independent experiments each performed in biological triplicate. age 50 Chromosome Y m onu c

10 r equen cy o m EYFP-AID en t h r c

DLD-1 CENP-A cells stably expressing H2B-mRFP were imaged Asterisks indicate significance by two-tailed Student’s t-test compared to c i

r 25 F h r wi t m c 2d after IAA addition. Scale bar, 5 μm. (d) DLD-1 cells as in (b) were untreated cells. **P=0.0019, NS = not significant. (h) Quantification of e C-H3 P 0 0 treated with combinations of dox and IAA for 24h and whole-cell extracts DLD-1 CENP-A cell growth rate with or without dox/IAA over a 9d period T 3 e W C-H were analyzed by immunoblotting for CENP-A. The predicted molecular performed in biological triplicate. Line represents linear regression analysis. A A Dox/IAA: - + - + C-H3 No rescu CENP- CENP- weight of CENP-A fused to an EYFP-AID tag is ~66 kDa. (e) Representative (i) Estimated doubling time calculated from h. (j) 5d CENP-A -rescued CENP-AWT CENP-AC-H3 immunofluorescent images of engineered DLD-1 cells treated with cells were subjected to propidium iodine staining followed by flow cytometry combinations of dox and IAA for 24h. Both CENP-AWT and CENP-AC-H3 analysis for DNA content with and without 6h treatment with 100 ng/ml c Estimated frequency of total cells with: rescues correctly localized to centromeres. Enlarged images of CENP-A nocodazole. Source data for g and h have been provided in Supplementary Rescue CENP-A Condition Chr. 4 micronuclei Chr. Y micronuclei staining following dox/IAA addition is shown below. ACA; anti-centromere Table 1. Wild-type ---- 0.09% (1 in 1,111) 0.35% (1 in 286) Dox/IAA (5d) 0.21% (1 in 476) 1.81% (1 in 55) C-H3 chimera ---- 0.22% (1 in 455) 0.95% (1 in 105) Dox/IAA (5d) 0.37% (1 in 270) 12.05% (1 in 8)

d e

i 2 3 8 R = 0.70

o c eus M) l o f ea r μ

l e f ( 6 2 e r onu c be r e r r

c 4 e t onu c i o m r r u m m 1 c i N

a m

en t 2 M c d i pe r 0 0 Dox/IAA: - + - + 1 2 3 4 5 6 7 8

CENP-AWT CENP-AC-H3 Number of Y centromere foci

f Characteristic: CENP-A CENP-AC-H3 Localization centromeric centromeric Cell proliferation rate 25±2 hrs 27±3 hrs Long-term clonal growth yes yes Cell cycle distribution normal normal Mitotic duration normal normal Karyotypic stability yes yes Y chromosome instability no yes

WWW.NATURE.COM/NATURECELLBIOLOGY 2 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARYSupplementary INFORMATION Figure 2

b a -Dox/IAA +Dox/IAA p=0.0013 p=0.0016 p=0.016

ll s )

100 n i e ei p=0.0079 l 40 p=0.0004 % c o f ( e f

o 20 75 e i Chromosome 4 l o m onu c r c o s i age 50 Chromosome Y m onu c

10 r equen cy o m en t h r c c i

r 25 F h r wi t m c e P 0 0 T 3 e W C-H A A Dox/IAA: - + - + No rescu CENP- CENP- CENP-AWT CENP-AC-H3 c Estimated frequency of total cells with: Rescue CENP-A Condition Chr. 4 micronuclei Chr. Y micronuclei Wild-type ---- 0.09% (1 in 1,111) 0.35% (1 in 286) Dox/IAA (5d) 0.21% (1 in 476) 1.81% (1 in 55) C-H3 chimera ---- 0.22% (1 in 455) 0.95% (1 in 105) Dox/IAA (5d) 0.37% (1 in 270) 12.05% (1 in 8)

d e

i 2 3 8 R = 0.70

o c eus M) l o f ea r μ

l e f ( 6 2 e r onu c be r e r r

c 4 e t onu c i o m r r u m m 1 c i N

a m

en t 2 M c d i pe r 0 0 Dox/IAA: - + - + 1 2 3 4 5 6 7 8

CENP-AWT CENP-AC-H3 Number of Y centromere foci

f Characteristic: CENP-A CENP-AC-H3 Localization centromeric centromeric Cell proliferation rate 25±2 hrs 27±3 hrs Long-term clonal growth yes yes Cell cycle distribution normal normal Mitotic duration normal normal Karyotypic stability yes yes Y chromosome instability no yes

Supplementary Figure 2 Induced Y centromere inactivation provokes treated as indicated by extrapolating the percentage of micronucleated Y chromosome missegregation into micronuclei. (a) DLD-1 cells were cells and the percentage of micronuclei containing either chromosome rescued with CENP-AWT or CENP-AC-H3 for 5d and the percentage of Y or 4. (d) Quantification of the number of Y centromere foci observed micronucleated cells were quantified by DAPI staining. Data represent in spontaneously-derived or induced micronuclei following 5d CENP- the mean ± SEM of n = 3 independent experiments (1,453–1,945 cells AC-H3 rescue. Data represent the mean ± SEM of n = 65 (WT, -dox/IAA), per condition). P-values indicate significance by two-tailed Student’s 76 (WT, +dox/IAA), 67 (C-H3, -dox/IAA) or 102 (C-H3, +dox/IAA) t-test compared to untreated cells. (b) DLD-1 cells were rescued with micronuclei. (e) Comparison between the number of Y centromere foci per CENP-AWT and CENP-AC-H3 for 5d and micronuclei were quantified for micronucleus and micronuclear diameter from 5d CENP-AC-H3-rescued the percentage harboring centromere Y or centromere 4 signal(s). Data cells. Data were compiled from n = 150 micronuclei pooled from 3 represent the mean ± SEM of n = 3 independent experiments (380–754 independent experiments, and means are indicated by the line. R2-value micronuclei) or the mean of 2 independent experiments (CENP-AWT, represents correlation of size and foci number by linear regression dox/IAA; 290 micronuclei). P-values indicate significance by two-tailed analysis. (f) Summary of cellular characteristics comparing untreated Student’s t-test compared as denoted. (c) Comparison between the (CENP-AEYFP-AID) cells with dox/IAA-treated (CENP-AC-H3) cells. Source frequency of cells with the specified chromosome in micronuclei when data for a and b have been provided in Supplementary Table 1.

WWW.NATURE.COM/NATURECELLBIOLOGY 3 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION Supplementary Figure 3 Supplementary Figure 4

2xRFP-NLS Merge w/ DAPI a a Micronuclear envelope disruption DAPI Chr. Y CEN-Y Merge Intact ei

l 100 (NLS+) NLS- age 75

onu c NLS+ r en t c c i

r 50 e m P

3d Dox/IAA

o f 25 Disrupted 0 (NLS-) -Dox/IAA +Dox/IAA b c

DAPI 3d Dox/IAA i 10 Merge o c R2=0.22

Y chromosome e f b Lamin B1 γH2AX w/ DAPI c 60 events e r 8 1.8 Y centromere o m )

Micronuclear H2AX signal intensity r

u . 6

a . 16 en t (

High DNA damage c y

t i 12 Y 4 en s

3.1 o f

n t i 8 Low DNA damage be r 2 u m e sc en t 4 No damage N uo r l 0 9.8 F 0 1 50 100 150 200 0 10 20 30 40 50 60 Individual micronuclei Number of Y fragments

1 µM PD-0332991 10 µM RO-3306 d e Asynchronous (CDK4/6 inhibitor) (CDK1 inhibitor) G2-phase G1-phase S-phase G1 G1 G2/M

Events S + Calyculin A + Calyculin G2/M G1 S S G2/M

DNA content

f Day -1 Day 0 Day 1 Day 2 Day 3 Day 4 Fig. 4b Plate cells +siRNA Immunoblot Fig. 4d-e Plate cells +Dox/IAA +siRNA FISH Fig. 4f Plate cells +siRNA FISH Fig. 4f Plate cells +Dox/IAA, +siRNA FISH Fig. 4g Plate cells +siRNA Count cell number Fig. 4h Plate cells +Dox/IAA +Inhibitor FISH

Supplementary Figure 3 Induced Y chromosome micronuclei share common experiments (166–294 total micronuclei). Scale bar, 5 μm. (b) DLD-1 features of spontaneously derived micronuclei including micronuclear CENP-AC-H3 cells treated with 5d dox/IAA were immunostained for the DNA envelope disruption and the acquisition of DNA damage. (a) DLD-1 CENP- damage marker γH2AX and nuclear envelopes with Lamin B1. Representative AC-H3 cells stably expressing 2xRFP-NLS treated with or without 5d dox/IAA images (scale bar, 5 μm) of micronuclei without and with varying degrees (experimentally vs. spontaneously derived micronuclei, respectively) were of detectable DNA damage signals are shown. (c) γH2AX fluorescent signal fixed and DAPI-stained. Representative images from dox/IAA-treated cells intensities from b were measured from 200 micronuclei (pooled from 3 and quantifications for micronuclear RFP compartmentalization are shown independent experiments) and individually plotted. a.u., arbitrary units. on the left. Data on the right panel represent the mean of 2 independent Source data for a and c have been provided in Supplementary Table 1.

WWW.NATURE.COM/NATURECELLBIOLOGY 4 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION Supplementary Figure 3 Supplementary Figure 4

2xRFP-NLS Merge w/ DAPI a a Micronuclear envelope disruption DAPI Chr. Y CEN-Y Merge Intact ei l 100 (NLS+) NLS- age 75 onu c NLS+ r en t c c i r 50 e m P

3d Dox/IAA o f 25 Disrupted 0 (NLS-) -Dox/IAA +Dox/IAA b c

DAPI 3d Dox/IAA i 10 Merge o c R2=0.22

Y chromosome e f b Lamin B1 γH2AX w/ DAPI c 60 events e r 8 1.8 Y centromere o m )

Micronuclear H2AX signal intensity r u . 6

a . 16 en t (

High DNA damage c y t i 12 Y 4 en s

3.1 o f n t i 8 Low DNA damage be r 2 u m e sc en t 4 No damage N uo r l 0 9.8 F 0 1 50 100 150 200 0 10 20 30 40 50 60 Individual micronuclei Number of Y fragments

1 µM PD-0332991 10 µM RO-3306 d e Asynchronous (CDK4/6 inhibitor) (CDK1 inhibitor) G2-phase G1-phase S-phase G1 G1 G2/M

Events S + Calyculin A + Calyculin G2/M G1 S S G2/M

DNA content

f Day -1 Day 0 Day 1 Day 2 Day 3 Day 4 Fig. 4b Plate cells +siRNA Immunoblot Fig. 4d-e Plate cells +Dox/IAA +siRNA FISH Fig. 4f Plate cells +siRNA FISH Fig. 4f Plate cells +Dox/IAA, +siRNA FISH Fig. 4g Plate cells +siRNA Count cell number Fig. 4h Plate cells +Dox/IAA +Inhibitor FISH

Supplementary Figure 4 Characterization of chromosome fragmentation with calyculin A, showing examples for G1-, S-, and G2-phase spreads. events and induction of premature chromosome condensation using G1-phase chromosomes appear as single chromatids, S-phase appears as calyculin A. (a) Representative example of interphase cells hybridized highly pulverized and abnormal nuclei (and excluded from quantitative to Y chromosome paint (green) and Y centromere (red) FISH probes analyses), and G2-phase appears as normal mitotic chromosomes with following 3d CENP-AC-H3 rescue. Scale bar, 10 μm. (b) Additional two distinguishable sister chromatids. Scale bar, 25 μm. (e) DLD-1 cells example of Y chromosome fragmentation event derived from 3d CENP- treated with 1 μM of the CDK4/6 inhibitor PD-0332991 (also known AC-H3-rescued cells. Scale bar, 10 μm. (c) Centromere and fragment as Palbociclib) or 10 μM of the CDK1 inhibitor RO-3306 for 24h were counts from Fig. 3f-g were cross-plotted per mitotic shattering event. subjected to propidium iodine staining followed by flow cytometry analysis Red line indicates linear regression analysis. (d) Representative image for DNA content. (f) Experimental schematic for panels shown in Fig. 4b, of DAPI-stained, metaphase-like spreads induced by 1h treatment d, e, f, g, and h.

WWW.NATURE.COM/NATURECELLBIOLOGY 5 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION

Supplementary Figure 5

a b Genomic Micronuclear ) Genomic Micronuclear h bp) 1000 % ( 1 2 3 4 5 6 ( e bo t z

i 750 100 h s wi t apped 500 en t

m 50 250 ead s ag m R r end s F 0 0 1 2 3 1 2 3 1 2 3 1 2 3

c Genomic DNA (n=3) Micronuclear DNA (n=3)

% 20 % 20 ], ], s s r 15 r 15 pa i pa i 10 10 dan t dan t r r 5 5 Di sc o Di sc o [ 0 [ 0 0 2 4 6 8 10 0 2 4 6 8 10 [Total pairs], % [Total pairs], %

Supplementary Figure 5 Paired-end sequencing information for each source and rises as the square of the concentration of total sequencing reads (see of DNA. (a) sizes (mean ± SD, n=1,000 reads) of sequencing Methods). Each dot represents a single chromosome from three independent fragments for each sample. (b) Percentage of sequencing reads in which genomic or micronuclear DNA samples, and the green dot indicates the Y both ends of a pair mapped to the reference genome following removal of chromosome. The curved line shows a predictive model of discordant pairs duplicate and mitochondrial reads. (c) The concentration of discordant that is described under the Methods section. Source data for a and b have sequencing reads for each chromosome follows a second order reaction been provided in Supplementary Table 1.

WWW.NATURE.COM/NATURECELLBIOLOGY 6 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION

Supplementary Figure 6

Supplementary Figure 5 Fig. 3h Supplementary Fig. 1d Supplementary Fig. 1f 80- 60-

α-CENP-A 15- α-CENP-A a b Genomic Micronuclear ) Genomic Micronuclear h 15- bp) 1000 % 15- 15- ( 1 2 3 4 5 6 ( e bo t z i 750 100 h s wi t apped 500 α-phospho-H3 (Ser 10) α-Total H3 40- α-GAPDH en t m 50 30-

250 40- ead s

ag m α-GAPDH R r 30- end s F 0 0 1 2 3 1 2 3 1 2 3 1 2 3 c Fig. 5b (left blot) Genomic DNA (n=3) Micronuclear DNA (n=3)

% 20 % 20 245-

], ], 190- 245- s s 190- 245- r r 46- 15 15 100- 135- 46- pa i pa i 32- 80- 32- 10 10 80- dan t dan t r r 5 5

Di sc o Di sc o α-LIG4 α-DNA-PKcs α-BRCA2 α-RAD51 α-GAPDH [ 0 [ 0 0 2 4 6 8 10 0 2 4 6 8 10

[Total pairs], % [Total pairs], % Fig. 5b (right blot)

135- 135- 100- 100- 80- 32-

α-LIG3 α-PARP-1 α-GAPDH

Supplementary Figure 6 Unprocessed film scans of all immunoblots with corresponding protein size markers.

WWW.NATURE.COM/NATURECELLBIOLOGY 7 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved. SUPPLEMENTARY INFORMATION

Supplementary Table Legends

Supplementary Table 1 Statistics source data.

WWW.NATURE.COM/NATURECELLBIOLOGY 8 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.