articles

An acetylated form of histone H2A.Z regulates chromosome architecture in Schizosaccharomyces pombe

© 2009 Nature America, Inc. All rights reserved.

Hyun-Soo Kim1,10, Vincent Vanoosthuyse2,10, Jeffrey Fillingham3, Assen Roguev4,5, Stephen Watt6, Thomas Kislinger7, Alex Treyer1, Laura Rocco Carpenter8, Christopher S Bennett8, Andrew Emili3,9, Jack F Greenblatt3,9, Kevin G Hardwick2, Nevan J Krogan4,5, Jürg Bähler6 & Michael-Christopher Keogh1 Histone variant H2A.Z has a conserved role in genome stability, although it remains unclear how this is mediated. Here we demonstrate that the fission yeast Swr1 ATPase inserts H2A.Z (Pht1) into chromatin and Kat5 acetyltransferase (Mst1) acetylates it. Deletion or an unacetylatable mutation of Pht1 leads to genome instability, primarily caused by chromosome entanglement and breakage at anaphase. This leads to the loss of telomere-proximal markers, though telomere protection and repeat length are unaffected by the absence of Pht1. Strikingly, the chromosome entanglement in pht1∆ anaphase cells can be rescued by forcing chromosome condensation before anaphase onset. We show that the condensin complex, required for the maintenance of anaphase chromosome condensation, prematurely dissociates from chromatin in the absence of Pht1. This and other findings suggest an important role for H2A.Z in the architecture of anaphase chromosomes. The basic repeating unit of chromatin is the nucleosome core particle: 146 base pairs (bp) of DNA wrapped around a core histone octamer composed of a (H3-H4)2 tetramer and two H2A-H2B dimers. One means of nucleosome specialization is the replacement of a major histone with a specific variant. These single-copy nonallelic isoforms are generally expressed throughout the cell cycle, and many enter nucleosomes through the action of specific deposition machineries1. Both major and variant histones can be further distinguished by the addition of small chemical moieties, including phosphorylation, acetylation, methylation and ubiquitylation2,3. H2A.Z is one of the most-studied histone variants. In metazoans H2A.Z is essential4, regulating chromosome stability5, gene activation6,7 and spermatogenesis8. The histone is subject to multiple N-terminal acetylations in Saccharomyces cerevisiae (Sc)9–11, Tetrahymena thermophila12 and metazoans13,14. Though the role(s) of these modi­ fications remains uncertain, defects in heterochomatin restriction11 and chromosome stability9 are observed when unacetylatable H2A.Z alleles are expressed as the sole source of the histone in budding yeast. A phenotype common to all H2A.Z-deficient species is genomic instability. This manifests in mammalian cells as chromatin bridges in anaphase5. Deletion of Sc H2A.Z (htz1∆) results in an increased rate of chromosome loss, sensitivity to microtubule destabilizing agents, and synthetic genetic interactions with components of the kinetochore and spindle checkpoint machineries15. Many of these phenotypes are shared by an unacetylatable htz1 mutant, suggesting a role for acetylation in genomic stability9. However, we still have

little mechanistic understanding of what leads to the chromosome segregation defect in these cells, though centromere organization, kinetochore attachment, cohesin recruitment and arm sister chrom­ atid cohesion appear normal9. To explain the role of H2A.Z in chromosome stability, we turned to the fission yeast Schizosaccharomyces pombe (Sp) as its three large chromosomes (3 × 3.5–5.7 Mb) can be more easily monitored during mitosis than those of budding yeast (16 × 230 kb–1.3 Mb). We show that Sp H2A.Z (pseudohistone 1, or Pht1) copurifies with a complex almost identical in composition to the Sc SWR complex (SWR-C, with this complex required for the insertion of Pht1 into chromatin. Chromatin-associated Pht1 is acetylated on its N terminus by the KAT5 acetyltransferase (Sp Mst1). This modification is essential for Pht1 function, with unacetylatable mutants phenocopying complete deletion of the histone variant in all analyses, including ­ genomescale genetic interaction and gene expression studies. In addition, we show that chromosome loss in pht1 mutants is primarily caused by broad architectural defects and can be suppressed by improving ­chromosome condensation. Consistent with this, we show by chromatin immunoprecipitation that the condensin complex is prematurely released from chromatin at anaphase in pht1∆ cells. RESULTS Pht1 is acetylated as a component of chromatin Sp Pht1 has four potentially acetylatable lysine residues on its N terminus: Lys5, Lys7, Lys12 and Lys16 (Fig. 1a). We synthesized

1Department

of Cell Biology, Albert Einstein College of Medicine, New York, New York, USA. 2Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh, Scotland, UK. 3Banting and Best Department of Medical Research, University of Toronto, Toronto, Canada. 4Department of Cellular and Molecular Pharmacology and 5The California Institute for Quantitative Biomedical Research, University of California–San Francisco, San Francisco, California, USA. 6Wellcome Trust Sanger Institute, Cambridge, UK. 7Ontario Cancer Institute, Toronto, Canada. 8Active Motif Inc., Lake Placid, New York, USA. 9Department of Molecular and Medical Genetics, University of Toronto, Toronto, Canada. 10These authors contributed equally to this work. Correspondence should be addressed to V.V. ([email protected]) or M.-C.K. ([email protected]). Received 5 January; accepted 10 September; published online 15 November 2009; doi:10.1038/nsmb.1688

1286

VOLUME 16  NUMBER 12  december 2009  nature structural & molecular biology

articles Figure 1  Chromatin-associated Pht1 is acetylated by Mst1. (a) Alignment of the H2A.Z N termini from Sc (Htz1, 3 8 10 14 YOL012C), Sp (Pht1, SPBC11B10.10c), Sc Drosophila melanogaster (Dm; H2Av, 5 7 12 16 15 kDa Anti-Pht1 Sp P028985), Mus musculus (Mm; NP_058030) 4 7 11 13 15 Anti-Pht1Ac 15 kDa and Homo sapiens (Hs; NP_002097). Dm Anti-H3 Pht1 is misannotated in the literature by 15 kDa Mm the addition of 32 N-terminal residues Anti-H3ac 15 kDa Hs (MILRHAPRVHESAFSLTHKTFAFCNCNNRFKM-). 37 kDa Anti-Rpn8 This would give a protein of 18 kDa rather (at 30 °C) than the 14 kDa observed. The acetylated Pht1.HA3 tag Mst1.HA3 mst1-L271P.HA3 residues on Sc Htz1 (Lys3, Lys8, Lys10 Tot Cyt Nuc Chr Tot Cyt Nuc Chr and Lys14) correspond to Sp Pht1 Lys5, Anti-Pht1 Pht1.HA3 Lys7, Lys12 and Lys16. A synthetic pht1-N�.HA3 Anti-Pht1Ac tetra-acetylated peptide covering this region Anti-HA (boxed) was injected into rabbits to create Anti-H3 Pht1.HA3 anti-Pht1Ac. (b) Pht1 is acetylated on the Anti-H3ac N terminus. Immunoblotting was performed Anti-Pht1Ac Rpn8 Pht1 Anti-Rpn8 * on strains expressing C-terminally HA3-tagged * (at 30 °C) Pht1 (or indicated mutants) at the endogenous Anti-H3 Pht1.HA3 pht1-4KQ.HA3 locus. Total Pht1 was detected by anti-HA Anti-Rpn8 Tot Cyt Nuc Chr Tot Cyt Nuc Chr (12CA5; Supplementary Table 3). * in Anti-HA anti-Pht1Ac panel refers to cross-reaction 30 °C 37 °C Ac Anti-Pht1 with H4ac. H3 and Rpn8 serve as loading WT controls. (c) The mst1-L271P allele is slow Mst1.HA3 Anti-H3 and ts. Spot tests are ten-fold dilutions Rpn8 mst1-L271P.HA3 Anti-Rpn8 * onto YES plates. (d) WCEs from mst1-L271P show a profound reduction in Pht1Ac without affecting total Pht1 or H3ac levels. H3 and Rpn8 serve as loading controls. (e,f) Pht1 acetylation by Mst1 is not required for assembly of the histone into chromatin. In f, strains express Pht1.HA3 or unacetylatable pht1-4KQ.HA3 at the endogenous locus. Cells were treated to form (Total, Tot) and fractionated into cytoplasm (Cyt), nucleus (Nuc) and chromatin (Chr). H3 and Rpn8 controls (*cross-reacting species) indicate efficient fractionation: the former is primarily localized in insoluble chromatin, the latter in soluble cytoplasm. W

T

P. H

A

3

d

e

© 2009 Nature America, Inc. All rights reserved.

U

nt ag -N � -4 KR -4 KQ W T

b

M st 1. H A m st 3 1L2 71

a

f

c

a peptide ­containing all four acetyllysines and immunized rabbits to create polyclonal anti-Pht1Ac. On immunoblots of Sp whole cell extracts (WCEs) this antibody recognized a protein of ­appropriate size for Pht1. This signal disappears in WCEs from pht1∆, -N∆ (deletion of the N terminus), or mutants where all four lysines were mutated to unacetylatable arginine or glutamine (Fig. 1b). Thus, the Pht1 N terminus is acetylated and we have a specific reagent for the modification. The primary acetyltransferase for Sc Htz1 is Esa1, the catalytic subunit of the NuA4 complex9–11. Esa1 is a member of the KAT5 family, with the most likely Sp homolog the essential protein Mst1 (ref. 16). We created a temperature-sensitive (ts) allele of Mst1 (mst1-L271P) that resulted in slow growth at the permissive temperature (25 °C) and was lethal at >34 °C (Fig. 1c). WCEs from mst1-L271P cells showed no appreciable change in total Pht1 levels but did show a profound reduction in Pht1Ac (Fig. 1d), indicating that Pht1 acetylation is Mst1 dependent. Cell fractionation showed that Pht1Ac is chromatin associated, though acetylation is not required for entry to this cellular compartment (Figs. 1e–f). Sc Htz1 associates with the SWR-C, containing the Swr1 ATPase, and incorporation of the histone into chromatin is markedly reduced in swr1∆ cells17–19. We subjected WCEs from Pht1.TAP to sequential affinity purification and identified the associated proteins by MS (Supplementary Table 1). Reciprocal tagging and purification of these factors delineates the Sp SWR-C, which is almost identical in composition to the Sc SWR-C: even the subunits shared between the Sp SWR-C and the Mst1-acetyltransferase complex are those shared by their Sc counterparts, the SWR-C and NuA4 (Fig. 2a). Western analysis with anti-Pht1Ac distinguished those subunits of the Sp SWR-C required for the efficient acetylation of the histone (Fig. 2b), most

likely because of inefficient assembly of the variant into chromatin in each background (Fig. 2c). Thus, a pathway first identified in Sc also operates in Sp: SWR-C inserts Pht1 into chromatin, where it is acetylated by Mst1. Acetylation is integral to Pht1 function To determine the relevance of N-terminal acetylation to Pht1 function, we compared the gene expression and genetic interaction profiles of various unacetylatable mutant alleles (pht1-N∆, -4KR, and -4KQ) to pht1∆. The transcription profiles of the unacetylatable mutants were almost identical to those of pht1∆ (for example, relative P value: ∆ to -4KR = 2e–195; Fig. 3a). A noteworthy observation was that almost all genes were modulated to some degree in pht1 mutants (Fig. 3b), suggesting that the histone acts as a general transcriptional regulator. By comparison, swr1∆ showed weaker changes in gene expression (Fig. 3b). As loss of the ATPase strongly reduces but does not completely ablate chromatin-associated Pht1 (Fig. 2c), it is likely that the remaining Pht1 is sufficient to mask most H2A.Zdependent phenotypes. We then used the Pombe Epistatic Mapper 2 (PEM-2) system to quantitatively analyze genetic interaction patterns20,21. To this end we individually crossed pht1∆, -N∆, -4KR, -4KQ and swr1∆ strains to a library of 2,161 nonessential Sp deletions (the Bioneer collection) and derived scores covering each negative (for example, synthetic sick or lethal) and positive (for example, suppression) genetic interaction using colony size as a quantitative readout20,22–24. Positive interactions enrich for factors that are in complex together or function in the same pathway20,22,23. Consistent with this, pht1∆, unacetylatable pht1 and swr1∆ each gave rise to positive genetic interactions in combination with deletion of all nonessential subunits of the SWR-C20 (Fig. 3c).

nature structural & molecular biology  VOLUME 16  NUMBER 12  december 2009

1287

articles

Rv

b2

b1

Yaf9

Swc4 Yaf9

Unique to SWR-C (Sc/Sp) Actin-related KAT5-C (Sc/Sp)

.0 2c �

Anti-Pht1 Ac

Anti-Pht1

RuvB-like ATPase Sc SWR-C only Sp SWR-C only

15 kDa

Anti-H3ac

15 kDa 37 kDa

Anti-Rpn8

2

4

–499 –275 49 283

1

3

–765 –510 –242 6 –3491

–2757

Untag 2

3

1053

WT 4

5

1

2

2044 2460

SPCC13B11.02c

Pht1.HA3 swr1�

3

4

5

1

2

3

msc1�

4

5

1

2

3

Anti-Tbp1

adh1 locus GFR

Input

adh1 locus GFR

KQ sw r1 � ph t1 � ph t1 -4 ph KR t1 -4 sw KQ r1 �

KR

c

-4

2.0 1.5

Expression

1.2

P values

0.1

pht1� /-4KR

2e–195

pht1� /-4KQ

4e–131

-4KR / -4KQ

2e–196

d

1.0 0.9 0.8

All genes

1,185 genes

e

swr1 yaf9 spac4h3.02c swc6 arp6 swc5 swc2 msc1

-4

c­ onfirming that they function in the same pathway. Indeed, of all mutants analyzed, the most highly correlated to pht1∆ were -N∆, -4KR and -4KQ, suggesting that N-terminal acetylation is critical for Pht1 ­function (Fig. 3e). Inspection of individual interactions showed that all unacetylatable pht1 alleles were synthetic, with deletions of factors involved in chromatin modification and/or remodeling (for example, COMPASS, RSC, SET3-C), transcription (for example, Mediator)

t1 ph

t1 �

t1

ph

ph

KQ sw r1 �

KR -4

b

t1

t1

ph

t1 �

ph

ph

10

5 adh1 locus GFR

-4

a

4

Anti-HA

This implies that preventing Pht1 acetylation disables the primary function of the SWR-C–Pht1 pathway in fission yeast. The genetic interaction profile of a particular mutant can be used as a high-resolution phenotype: comparison of this profile to those generated from other mutants can identify functionally related factors20,22,23. When compared to data from >100 genetic screens (data not shown), pht1∆ and swr1∆ were highly correlated (Fig. 3d),

Normalized intensity

1.0

pht1�

0.8 -4KQ

0.6 swr1� pht1� pht1-N� pht1-4KR pht1-4KQ

–3 –2 –1 0 1 2 3

Negative 0.7 interactions 0.6 (synthetically sick/lethal) 0.5

CC = 0.891 pht1-N� -4KR swr1�

0.4 0.2 0 –0.2

Positive interactions (suppressive/ epistatic)

–0.4 –0.4 –0.2

0

0.2 0.4 swr1�

0.6

0.8

1.0

f

Isk1 dad5 spc34 dad1 cut8 spn3 ppk21

ssu72 ash2 swd3 set3 raf1 cph1 hrp3 arp42 pmc2 pmc5 rsc1

Chromatin remodeling/ Cytokinesis CC = 0.919 Figure 3  Unacetylatable pht1 mutants phenocopy pht1∆. transcription -4KQ 1.0 (a) Panel compares genes whose expression is induced -4KR 0.8 pht1-N� (red) and repressed (blue) (by a factor of at pht1� 0.6 swr1� least 1.5 in at least one condition) as indicated by swr1� pht1� 0.4 whole-genome array data from each strain. Relative pht1-N� pht1-4KR P values for expression overlaps among induced genes 0.2 pht1-4KQ between strain pairs are as indicated. (b) Heat map of 0 Mediator DASH-C COMPASS induced (yellow) and repressed (blue) genes in each –0.2 –3 –2 –1 0 1 2 3 mutant. Left, 1,185 genes are changed by a factor of –0.4 Negative Positive at least 1.5 in at least one mutant background. Right: –0.4 –0.2 0 0.2 0.4 0.6 0.8 1.0 interactions interactions (synthetically (suppressive/ all genes on the array. (c) Unacetylatable pht1 mutants pht1� sick/lethal) epistatic) display positive genetic interactions with deletions of SWR-C subunits. (d) Correlation coefficient (CC) plot comparing the genetic interactions of 101 query mutants (including pht1∆, -N∆, -4KR, -KQ and swr1∆) mated against a library of 2,161 nonessential deletions. Red dots indicate the CC of each genetic screen to the mutants on the x or y axis. (e) CC plot of pht1∆ versus pht1-4KQ. (f) pht1∆, -N∆, -4KR, -KQ and swr1∆ share a large number of synthetic genetic interactions, including with members of the COMPASS, RSC, SET3-C, Mediator and DASH complexes. pht1-4KQ

© 2009 Nature America, Inc. All rights reserved.

1

5

366 609 adh1+

1

1 kb

SPCC162.01c

15 kDa

Anti-H3

Tra2

c

15 kDa

pht1�

Swr1

Rv

Arp6

Arp4

Swc4

Msc1

Arp6

Act1

Arp4

Swr1

Swc5 Swc6

Swc2 SPAC 4H3.02c Act1

Swc7 R Rv vb1 b2

Swc5 Swc6

Figure 2  Pht1 is inserted into chromatin by the SWR-C. (a) The SWR-C is highly conserved from Sp to Sc. Multiple subunits of each complex were C-terminally tagged for TAP and associated factors identified by MS17 (Supplementary Table 1). In this schematic, location does not indicate direct interaction. (b) SWR-C components are individually required for the efficient acetylation of Pht1, suggesting chromatin-insertion defects. Total Pht1 levels are also reduced in many of these mutants. Immunoblotting of H3, H3ac and Rpn8 serve as loading controls. (c) Pht1 is inefficiently assembled into chromatin in swr1∆, and to a lesser extent in msc1∆. Schematic depicts the neighborhood of constitutively expressed adh1+. Black boxes depict the location of ChIP primers (Supplementary Table 5) relative to the start codon (+1). Pht1.HA3 was detected by anti-HA. Appropriate localization of Tbp1 at the adh1+ promoter confirms sample integrity. In these duplex reactions, upper band (adh1 locus) is the specific primers numbered in the schematic; lower band is a nontranscribed gene-free region (GFR) included as a loading control. Lower panel (Input) is used to normalize the PCR amplification efficiency of each primer pair.

T ph t1 sw � r1 sw � c2 sw � c5 ar � p6 ya � f9 � m sc 1� sp ac 4h 3

Swc2 Swc3

b

S. pombe

S. cerevisiae

W

a

1288

VOLUME 16  NUMBER 12  december 2009  nature structural & molecular biology

articles

t ph 1� t1 -N � ph t1 -4 ph K R t1 -4 KQ cl r4 �

a. pht1�

b. pht1�

3 µm

c. pht1�

3 µm

d taz1-GFP

100 80 60

WT

40

3 µm

20 °C

f Gar2-GFP

Nucleolus DNA

merge

WT

12 6 5 4 3

pht1�

2 1.6

20 0

30 °C

kb

1

WT pht1� clr4�

Pht1Ac

pht1�

3 µm

g

0.5

EcoRI digest

pht1�

h

Figure 4  Lack of leads to chromosome segregation defects Left arm of chromosome 1, 80 kb from telomere in anaphase. (a) The percentage (mean ± s.d.) of late anaphase cells sod2-GFP DNA merge with chromosome segregation defects (see succeeding panels) was 3.5 visually scored. All strains were grown in YES at 30 °C. (b) Kinetochore WT n = 1,800 3.0 pht1� segregation was monitored by distribution of Nuf2-GFP 50. Three sgo2� chromosome mis-segregation defects (arrowed) are distinguished 2.5 3 µm in pht1∆ anaphase cells: lagging, entanglement, and entanglement pht1� 2.0 leading to breakage. (c) The primary chromosome segregation defect in n = 1,310 1.5 pht1∆ (entanglement) is distinct from that in heterochromatin-deficient clr4∆ (lagging). Bars are mean ± s.d. (d) Telomere segregation was 1.0 n = 1,900 3 µm pht1� monitored by distribution of Taz1-GFP29. In the example two individual n = 2,550 0.5 telomeres (two Taz1 spots) are entangled in the middle of a pht1∆ n = 1,100 n = 900 n = 850 0 anaphase cell, leading to stretched chromosome arms. (e) Pht1 0 500 1,000 1,500 2,000 (in contrast to Taz1) does not regulate telomere repeat length. Distance of the marker from centromere 3 µm (f) Nucleolar segregation was monitored by distribution of Gar2-GFP 51. pht1� In many pht1∆ cells the nucleolus appears stretched or fragmented. (g,h) Chromosome marker loss in pht1∆ cells increases with distance from the centromere. GFP-marked loci locations (cen2-lacO, 5 kb from CEN2; ade3-lacO, 1,350 kb from TEL1-l; sod2-lacO, 80 kb from TEL1-l (ref. 30)) were used to monitor and quantify chromosome segregation defects. n represents the number of binucleate cells counted for each marker in each background. Percentage of mis-segregation of the marker

© 2009 Nature America, Inc. All rights reserved.

e

Telomeres DNA merge

W T ph ta t1 � z1 W � phT ta t1� z1 �

Percentage of anaphase defects being lagging chromosomes

c

KINETOCHORES merge DNA

nuf2-GFP

ph

Lagging Breakage

Entanglement

b 50 45 40 35 30 25 20 15 10 5 0

W T

Percentage of anaphase cells with chromosome segregation defects

a

and chromosome segregation and/or cytokinesis (for example, cut8, DASH complex) (Fig. 3f). These are reminiscent of the synthetic interactions displayed by Sc htz1∆15,17, further suggesting that there is strong conservation of H2A.Z function in each organism. Chromosome loss in pht1 is caused by entanglement at anaphase Knockout or depletion of H2A.Z in Sc15 or mammalian cells5 leads to increased rates of chromosome loss. This phenotype was also observed if any component of the Sp Pht1Ac pathway was disrupted, including mutants in swr1 (and msc1), pht1 (pht1∆, -4KR or -4KQ) or mst1 (refs. 16,25,26) (Supplementary Table 2). One possible explanation for chromosome instability is disruption of the centromere 27. However, as in htz1∆ cells9, centromere structure and function appeared normal in pht1 cells (Supplementary Fig. 1). Cytological analysis of individual Sp cells revealed a more than eight-fold increase (relative to wild type (WT)) in the number of pht1∆ cells with anaphase chromosome segregation defects (Fig. 4a). Three specific categories of anaphase defects were distinguished using the kinetochore marker Nuf2-GFP: (i) lagging chromosomes, in which the mis-segregating chromatin contains at least one kinetochore; (ii) chromosome entanglement, in which the missegregating chromatin is stretched between the spindle poles but contains no kinetochore; and (iii) entanglement leading to breakage, in which broken pieces of chromatin with no kinetochore lag on the spindle (Fig. 4b). The primary segregation defect in pht1∆ cells was entangle­ment (> 80% of defective anaphases). This was clearly distinct from the primary anaphase segregation defect in ­heterochromatin-­deficient clr4∆ cells (> 90% lagging chromosomes) (Fig. 4c). Lagging ­chromosomes result from merotely, a defect in which a single kinetochore attaches to microtubules emanating from opposite spindle poles28. Thus, to a large extent, kinetochoremicrotubule attachments appear normal in pht1 cells, supporting our observation from chromatin immuno­precipitation (ChIP)

s­ tudies of a WT-like centromere in pht1 mutants (Supplementary Fig. 1c). Consistent with this result, pht1 cells did not activate the spindle checkpoint, nor did they rely on spindle-checkpoint genes for survival (data not shown). Chromosome entanglement in pht1 cells was particularly obvious when we followed telomere segregation at anaphase. To do so, we used the telomere-binding protein Taz1-GFP as a marker. In a normal mitosis, six Taz1-GFP spots (identifying the left and right telomeres on the three Sp chromosomes) migrate to opposite poles with the bulk of DNA. However, in pht1 cells we often observed two or more Taz1-GFP foci entangled in the center, leading to stretched chromosome arms (Fig. 4d). One possible explanation for this entanglement is a defect in telomere function comparable to that observed in cells lacking the protection factor Taz1 (ref. 29). However, many of the phenotypes of taz1∆ were not shared by pht1∆: for example, cold sensitivity, size of the chromatin bridge, presence of Rad22 foci, and rqh1-SM suppression (Supplementary Fig. 2). In addition, and most noteworthy, although telomere-repeat length was greatly increased in taz1∆, it was comparable to WT in pht1∆ (Fig. 4e). To quantify how frequently the entanglement in pht1 cells leads to chromosome loss, we employed a series of strains containing: (i) lac­Operator (lacO) repeats integrated at different chromosomal locations, and (ii) LacI-GFP, the fluorophore-marked lacO binding protein30. Unexpectedly, marker loss in mitotic pht1∆ cells increased with distance from the centromere, suggesting that they generally lost broken pieces of chromosomes rather than whole chromatids (Figs. 4f–h). This is in sharp contrast to sgo2∆ cells, which are unable to correct chromosome biorientation defects31,32. In sgo2∆ cells marker loss was similar irrespective of chromosome position, suggesting the loss of whole chromatids (Fig. 4h). Together these observations suggest that the primary role of Pht1 in chromosome transmission is to maintain overall chromosome architecture rather than to regulate the function of a specific region (such as the centromere or telomere).

nature structural & molecular biology  VOLUME 16  NUMBER 12  december 2009

1289

articles a

Pht1 has a role in chromosome architecture and/or compaction Chromosomes condense to a folded rod-shaped structure upon mitotic entry. As the chromatids are pulled to the spindle poles in early anaphase, the chromosome arms trail behind the centromeres, forming a distinctive ‘arrowhead’ structure. This was lost in > 50% of cells lacking pht1 (Fig. 5a). The occurrence of such a generalized defect further supports the idea that Pht1 regulates overall chromosome architecture rather than that of specific loci. The primary chromosome segregation defect in pht1 cells, chromo­some entanglement despite accurate centromere segregation (Fig. 4), is reminiscent of that observed in mutants of condensin, a five-subunit complex instrumental to the architecture and segregation of chromosomes in mitosis33–36 (Supplementary Fig. 3a). We thus speculated that pht1 mutants may misregulate chromosome condensation during anaphase. If so, it may be possible to suppress their defect by forcing chromosome hypercondensation. To this end we used the cold-sensitive tubulin mutant, nda3-KM311, that fails to assemble microtubules at the restrictive temperature (20 °C), leading to a spindle-dependent checkpoint arrest37. This prolonged early mitotic arrest induces chromosome hypercondensation 37, and when shifted to the permissive temperature (32 °C) these cells enter a synchronous anaphase 31,37. This prolonged mitotic arrest specifically rescued the chromosome segregation defect of pht1∆ but not taz1 cells (Fig. 5b). Arresting pht1∆ cells in S-phase with hydroxyurea (HU), which depletes nucleotide pools, had no effect on the chromosome segregation defect, showing that merely prolonging the cell cycle was not sufficient to rescue pht1 defects (Fig. 5b). 1290

0 25 50 75 10 0 12 5 15 0

50 75 10 0 12 5 15 0 17 5

0 25 50 75 10 0 12 5 15 0 17 5

75 10 0 12 5 15 0 17 5

0 25 50

0 25 50 75 100 125 150 175 0 25 50 75 100 125 150 175

0 25

Relative fold enrichment

Percentage of anaphase cells where chromosome do not form arrowhead structure

Septation index (%)

c

Percentage of anaphase cells with chromosome segregation defects

b

© 2009 Nature America, Inc. All rights reserved.

d

Figure 5  Pht1 plays a role in chromosome H3K9me2, WT Cut3.HA3, WT architecture and compaction. (a) pht1∆ H3K9me2, pht1� Cut3.HA3, pht1� WT pht1� 1.4 1.0 70 (and -4KR or -4KQ; not shown) cells frequently 1.2 60 0.8 1.0 lose the ‘arrowhead’ structure of segregating 50 0.6 0.8 40 chromosomes in anaphase, indicating 0.6 0.4 30 0.4 20 a disruption of chromatin architecture 0.2 0.2 10 (mean ± s.d.). In the example, each anaphase 0 0 0 5 WT pht1� (WT and pht1∆) is at roughly the same stage, 17 imr1 otr1 6.5 µm between the poles. (b) The chromosome WT pht1� 30 °C Cycling (20 °C) segregation defects of pht1∆, but not taz1∆, can Time after Release from Release from HU block mitotic block release (min) be rescued by chromosome hypercondensation C3.5 35 30 Cut3 (anti-HA) induced by prolonged mitotic arrest. WT, GFR 30 25 C3.5 25 pht1∆ or taz1∆ cells in the background of 20 Anti-H3K9me2 GFR cnt1 20 15 Nda3 (blue) or a cold-sensitive tubulin C3.5 15 Input 10 GFR 10 mutant nda3-KM311 (red) were placed at 5 5 l3 Cut3 (anti-HA) the restrictive temperature (20 °C, 6 h), where 0 0 GFR WT pht1� taz1� WT pht1� nda3-KM311 failed to assemble microtubules l3 Anti-H3K9me2 GFR imr1 30 °C 34 °C 37 °C and underwent spindle-dependent checkpoint l3 WT Input arrest. Cells were released into anaphase at the GFR pht1� permissive temperature (32 °C) and scored for O2 cut3-477 Cut3 (anti-HA) GFR chromosome segregation defects. Arresting with pht1� cut3-477 O2 otr1 Anti-H3K9me2 cut14-208 HU had no effect on the phenotype. (c) pht1∆ GFR pht1� cut14-208 O2 partially rescued mutants in three subunits Input cnd2-1 GFR of condensin (each SMC and the kleisin: pht1 �cnd2-1 N1 Cut3 (anti-HA) rad21-K1 cut3-477, cut14-208 and cnd2-1), but is GFR pht1� rad21-K1 N1 synthetic with a mutant in cohesin (kleisin: Anti-H3K9me2 GFR rDNA WT rad21-K1). The ability to rescue condensin N1 Input set1� was not shared by deletions of the Set1 GFR gcn5� 100 100 methyltransferase or Gcn5 acetyltransferase. 75 75 cut14-208 50 50 25 25 Spot tests were ten-fold dilutions onto YES set1� cut14-208 0 0 gcn5� cut14-208 plates. (d) WT or pht1∆ (additionally containing Cut3.HA3 and cdc25-22) were arrested in G2 and released into the cell cycle (with samples taken every 25 min as in Online Methods). Septation index confirmed that each population completed mitosis by 125 min (peak septation, shaded in gray). ChIP was used to monitor condensin and H3K9me2 at various locations, including the Chr I centromere (for primer schematic see Supplementary Fig. 1c) and rDNA. Whereas H3K9 dimethylation at imr1 and otr1 followed similar kinetics in WT and pht1∆, Cut3.HA3 dropped prematurely at all locations in the absence of the histone variant (see also Supplementary Fig. 3d). “Relative fold enrichment” in the ChIP quantitation graphs is the ratio between the specific signal at each location and the respective input.

A functional relationship would be indicated by genetic inter­action, so we tested for that between pht1 and condensin. Unacetylatable pht1 or pht1 each raised the restrictive temperature of ts alleles of three complex subunits: cut3-477 (smc4-S1147P), cut14-208 (smc2-S861P) and cnd2-1 (cnd2-A114T) (Fig. 5c and Supplementary Fig. 3b). This partial rescue was specific, as pht1 was synthetic with rad21-K1, a mutant in the ­condensinrelated ­complex cohesin, which holds sister chromatids together before anaphase onset. This suppression is unlikely to be mediated through indirect ­transcriptional effects (for example, increased expression of condensin) because the mRNA levels (as determined by gene expression ­microarray) of all tested complex subunits and known regulators (for example, Cut17, Ark1, Top2, Fin1, Pim1, Pic1, Acr1, Nuc1) in WT cells were comparable to those in pht1∆ and unacetylatable pht1 cells (Supplementary Fig. 3c and data not shown). In addition, the partial rescue was not observed with deletion of two other transcriptional regulators: the Set1 methyl­transferase or Gcn5 acetyltransferase (Fig. 5c). Our results are consistent with a model where an acetylated form of Pht1 regulates condensin loading and/or localization in mitosis. To test this, we arrested WT or pht1 cells expressing Cut3 C-terminally tagged with the hemagglutinin YPYDVPDYA peptide (Cut3.HA3) at the G2-M boundary, and then monitored ­condensin occupancy kinetics through a synchronous mitosis (Fig. 5d). To first ensure that mitotic progression in WT and pht1∆ cells followed similar ­kinetics, we examined the peak septation index and H3K9me2 in each ­population. A newly formed septum indicates the completion of mitosis and can be visualized by specific staining with Calcofluor (see Online Methods). H3K9me2 is markedly reduced at ­centromeric repeats (otr) during mitosis, increasing as cells enter G1/S38. Each

VOLUME 16  NUMBER 12  december 2009  nature structural & molecular biology

a

M phase

S phase Prophase

Metaphase

Anaphase

Telophase

H2A.Z�

WT

Replication

b

c

M phase Metaphase

M phase Prophase

Anaphase

Metaphase

Anaphase

condensinmut H2A.Z�

Condensinmut

Prophase

d

Regulator of Chromosome Architecture (RCA)

Condensin

Compaction

Maintenance

Prophase On chromatin

Figure 6  H2A.Z plays a role in higher-order chromosome architecture. (a). Upper row: the localization and association of condensin with chromosomes is tightly regulated. The complex is primarily (but not exclusively) found in the cytoplasm through interphase, and imported into the nucleus at prophase for chromosome loading, with levels peaking at anaphase52,53. Lower row: the pathway in H2A.Z∆ cells. Condensin loads normally but prematurely dissociates (Fig. 5d). This explains the poorly resolved structure observed in anaphase (Fig. 5a), which likely leads to chromosome entanglement and loss (Fig. 4). (b) Sister chromatids in condensin mutants are ‘fuzzy’ and resolve poorly at prophase. The sisters remain connected by chromatin bridges as they pull apart in anaphase (see Supplementary Fig. 3a). (c) The rescue by pht1∆ (and unacetylatable mutants) of various chromosome architecture mutants (condensin and Topo II; Fig. 5c and Supplementary Figs. 3b and 4) indicates that a partially compensatory condensed topology exists in pht1∆ cells. (d) Model separates mitotic chromosome condensation and architecture into two stages: initial compaction and maintenance. The first stage is dependent on the poorly characterized activity (or factor) RCA; the second, on the condensin complex and topoisomerase II. H2A.Z may have a dual regulatory role: somehow opposing RCA, yet stabilizing the association of condensin with chromatin.

Metaphase

Anaphase

Condensin Topoll p

p

pattern was indistinguishable between WT and pht1∆ cells (Fig. 5d). When we examined condensin recruitment, Cut3.HA3 ­initially (≤ 75 min) followed similar kinetics at the centromere (cnt, imr and otr) and ­ribosomal DNA of WT and pht1∆ cells (Fig. 5d and Supplementary Fig. 3d). However, ­significant ­ differences were then observed: Cut3.HA3 levels ­continued to increase in WT cells, ­peaking at 100 min before dropping at 125 min (the latter corresponding to the peak septation index time point; Fig. 5d). However, in pht1∆ condensin delocalization began by 100 min, indicating the premature ­dissociation of the complex from chromatin. This likely explains many of our observations in these cells: loss of the anaphase arrowhead structure leading to chromosome entanglement and the loss of ­telomere­proximal regions (Fig. 5). DISCUSSION In this work we identify and characterize a previously unknown role for the histone variant H2A.Z (Sp Pht1) in chromosome ­architecture at anaphase. In the absence of Pht1, chromosomes frequently ­entangle in anaphase, which can lead to breakage and loss, particularly of telomere-proximal regions. We provide evidence that chromosome entanglement in pht1∆ is most likely due to premature dissociation of the condensin complex in anaphase. We also demonstrate that the factors involved in the chromatin loading and acetylation of H2A.Z are highly conserved in Sc and Sp. The last common predecessor of these yeasts was ~380 million years ago (mya)39; by comparison, the last common predecessor of the entire mammalian class existed ~165 mya40, whereas the primate line split to humans and gorillas ~8 mya41.

Stabilize

p

RCA

Destabilize

Ac

© 2009 Nature America, Inc. All rights reserved.

articles

H2A.Z p

p Repo-Man PP1

CDK

Repo-Man PP1

CDK

We propose that because the pathways regulating H2A.Z localization, modification and function are so well conserved, the role of the histone variant in chromosome architecture will be equally widespread. Extensive conservation of the SWR-C Comprehensive proteomic analyses identify two subunits in the Sp SWR-C not seen in its Sc counterparts Msc1 and SPAC4H3.02c (ref. 42) (Fig. 2a and Supplementary Table 1). Both deletions are epistatic with pht1 (ref. 20) (Fig. 3c), further suggesting a functional relationship. Msc1 was originally identified as a multicopy suppressor of cells defective for checkpoint kinase Chk1 (Rad27) function43. Recent work has found an epistatic chromosome loss phenotype in pht1∆ and msc1∆, and has suggested that Msc1 is upstream of Pht1 in this ­pathway26, a relationship our data supports (for example, Fig. 2c). Msc1 was recently shown to interact with the Mst1 acetyltransferase by yeast two-hybrid analysis16. Despite this, any physical interaction between Mst1 and Msc1 is likely to be transient, as Msc1.TAP purification did not identify Mst1 (or any nonshared subunit of the Swr1 and Mst1 complexes; Supplementary Table 1), and unique members of the Mst1-C do not copurify Msc1 (data not shown). However, the

nature structural & molecular biology  VOLUME 16  NUMBER 12  december 2009

1291

articles

© 2009 Nature America, Inc. All rights reserved.

link between the Swr1 and Mst1 complexes is interesting, and may suggest that Msc1 directly recruits Mst1 to sites of Pht1 integration. N-terminal acetylation is integral to Pht1 function The SWR-C is required for the assembly of Pht1 into chromatin, where it is acetylated by Mst1 (Figs. 1 and 2). This relationship between the ATPase (Swr1), histone variant (H2A.Z) and KAT5-­family acetyltransferase (Mst1) appears to be widely conserved 9,15,44–47. Unacetylatable pht1 phenocopies pht1∆ throughout this work but is perhaps most noteworthy in large-scale gene expression and genetic analyses (Fig. 3). This strongly suggests that acetylation of the histone is integral to its function. Based on Sc Htz1, it is likely that all four lysine residues in the Pht1 N terminus are modified10,11. However, our Pht1Ac antibody was raised to a tetra-acetylated peptide (Fig. 1a), and the unacetylatable pht1 used throughout this work has mutations at all four N-terminal lysines, so these individual modifications are as yet unconfirmed. mst1-L271P reduces Pht1Ac below the thres­ hold of detection (Fig. 1d), which suggests that we have identified the major enzyme for this modification. In this regard, we note that mst1 mutants also show increased rates of chromosome loss16, likely due in part to reduced Pht1Ac. Pht1 regulates chromosome architecture at anaphase Knockout or depletion of H2A.Z in Sc15, Sp25,26 or mammalian cells5 leads to increased rates of chromosome loss. An acetylated form of H2A.Z mediates chromosome stability role in both Sc9 and Sp (this work), strongly suggesting that this will prove to be the case in other organisms. Directly monitoring chromosome segregation in individual Sp cells allowed us to show that lack of Pht1 induces chromosome arm entanglements in anaphase that can lead to chromosome breaks (Fig. 4). Furthermore, Pht1 is required for the stable association of condensin with chromatin through anaphase (Fig. 5d), and the chromosome entanglement in pht1∆ cells can be rescued by a pre-anaphase arrest where chromosomes hypercondense (Fig. 5b). Thus, the lack of Pht1 interferes with chromosome architecture in anaphase. However, it also improves the viability of mutants that regulate this process, such as those in condensin (cut3-477, cut14-208, cnd2-1) or topoisomerase II (top2-191) (Fig. 5c and Supplementary Fig. 4). To resolve this apparent contradiction, we propose that Pht1 actually plays a dual role in mitotic chromosome architecture (see below and Fig. 6). Mitotic defects in a chicken condensin mutant primarily occur in anaphase: chromosomes prematurely lose their compact organization as they move to the poles48. Thereafter, individual chromatids can no longer be distinguished and prominent chromatin bridges are visible in > 90% of cells (for example, Fig. 6b and Supplementary Fig. 3a). These phenotypes can be overcome if Repo-Man, a targeting subunit for protein phosphatase 1 (PP1), is unable to direct the phosphatase to chromosomes in anaphase48. In WT cells Repo-Man is subject to CDK-dependent phosphorylation(s) to abrogate chromosome binding at early mitosis when CDK levels are high. Chromosome compaction at this stage is only slightly affected in condensin mutants, prompting the proposal that an as-yet-uncharacterized activity, regulator of chromosome architecture (RCA), drives initial condensation. RCA is then inhibited in a process dependent on Repo-Man and PP1 after CDK levels fall at anaphase onset and condensin steps in to stabilize chromosome architecture until mitosis completes48 (all modeled in Fig. 6d). The above model suggests that there are at least two steps in chromo­ some condensation. The first is RCA-dependent compaction to the characteristic X-shaped mitotic chromosome. The second is the condensin-dependent maintenance of a robust architecture that can 1292

withstand the pulling forces of microtubules in anaphase. Our data suggests that Pht1 regulates both steps: promoting the inhibition of RCA and stabilizing the association of condensin. As above, artificial maintenance of RCA activity in anaphase by preventing the loading of PP1 to chromatin partly rescues the chromosome segregation defects of chicken smc2 mutants48. This is reminiscent of the genetic inter­actions between pht1 and condensin (for example, Fig. 5c). It is highly likely that RCA is conserved in Sp, as deletion of the fission yeast PP1 ortholog that localizes on chromatin (PP1Dis2), also partly rescues cut3-477 (Supplementary Fig. 4b). Finally, we have recently shown that Sc Bud14, a regulator of Sc PP1 (Glc7), is required for the efficient loading of Htz1 onto chromatin (and subsequent acetylation of the histone variant)49. This suggests that H2A.Z could act downstream of PP1 to inhibit RCA in anaphase (Fig. 6d). Further studies are underway to test this model. Methods Methods and any associated references are available in the online version of the paper at http://www.nature.com/nsmb/. Accession code. Microarray data have been deposited at EMBL EBI ArrayExpress with accession code E-TABM-842. Note: Supplementary information is available on the Nature Structural & Molecular Biology website. Acknowledgments We thank R. Allshire (Univ. of Edinburgh), M. Bühler (Friedrich Miescher Institute for Biomedical Research), J. Cooper (Cancer Research UK), D. Finley (Harvard Medical School), S. Forsburg (Univ. of Southern California, Los Angeles), K. Gull (Univ. of Oxford), C. Hoffmann (Boston College), J. Kanoh (Kyoto Univ.), R. Maraia (NIH), D. Moazed (Harvard Medical School), T. Nakamura (Univ. of Illinois at Chicago), F. Neumann (Rockefeller Univ.), P. Nurse (Rockefeller Univ.), M. O’Connell (Mount Sinai School of Medicine), J. Partridge (St. Judes Children’s Research Hospital), M. Yanagida (Kyoto Univ.) and the Yeast Genome Resource Center (Osaka City Univ.) for the generous supply of antibodies and yeast strains (detailed in Supplementary Tables 3 and 4). We also thank G. Zhong, S. Chandran, T. Punna and M. Shales for technical support. Finally, we are grateful to G. Ingram for expertise with the Telomere Repeat Length assay. Work in the J.B. lab is funded by Cancer Research UK, T.K. lab by a start-up grant from the Ontario Cancer Institute, K.G.H. lab by a program grant from the Wellcome Trust and M.-C.K. lab by an NCI Cancer Center Support grant to Albert Einstein College of Medicine (2P30CA013330) and the Speaker’s Fund for Biomedical Research: Toward the Science of Patient Care, awarded by the City of New York. AUTHOR CONTRIBUTIONS H.-S.K. was responsible for the data in Figure 1 (with A.T. contributing Fig. 1c), Figure 2 (with MS help from J.F., T.K., A.E. and J.F.G.), Figures 5c and 5d and Supplementary Figures 1 and 3. V.V. (with help from K.G.H.) was responsible for the data in Figures 4 and 5a–c and Supplementary Figures 2, 3a and 4. S.W. and J.B. performed and analyzed the microarrays in Figures 3a and 3b; A.R. and N.J.K. performed and analyzed the genetic screens in Figures 3c–f. L.R.C. and C.S.B. created the antibodies to Pht1 used in Figure 1. H.-S.K., V.V. and M.-C.K. planned experiments, analyzed the data and wrote the manuscript. Published online at http://www.nature.com/nsmb/. Reprints and permissions information is available online at http://npg.nature.com/ reprintsandpermissions/. 1. Henikoff, S., Furuyama, T. & Ahmad, K. Histone variants, nucleosome assembly and epigenetic inheritance. Trends Genet. 20, 320–326 (2005). 2. Latham, J.A. & Dent, S.Y.R. Cross-regulation of histone modifications. Nat. Struct. Mol. Biol. 14, 1017–1024 (2007). 3. Millar, C.B. & Grunstein, M. Genome-wide patterns of histone modifications in yeast. Nat. Rev. Mol. Cell Biol. 7, 657–666 (2006). 4. Faast, R. et al. Histone variant H2A.Z is required for early mammalian development. Curr. Biol. 11, 1183–1187 (2001). 5. Rangasamy, D., Greaves, I. & Tremethick, D.J. RNA interference demonstrates a novel role for H2A.Z in chromosome segregation. Nat. Struct. Mol. Biol. 11, 650–655 (2004).

VOLUME 16  NUMBER 12  december 2009  nature structural & molecular biology

© 2009 Nature America, Inc. All rights reserved.

articles 6. Leach, T.J. et al. Histone H2A.Z is widely but non-randomly distributed in chromosomes of Drosophila melanogaster. J. Biol. Chem. 275, 23267–23272 (2000). 7. Rangasamy, D., Berven, L., Ridgway, P. & Tremethick, D.J. Pericentric heterochromatin becomes enriched with H2A.Z during early mammalian development. EMBO J. 22, 1599–1607 (2003). 8. Greaves, I.K., Rangasamy, D., Devoy, M., Marshall Graves, J.A. & Tremethick, D.J. The X and Y chromosomes assemble into H2A.Z, containing facultative heterochromatin, following meiosis. Mol. Cell. Biol. 26, 5394–5405 (2006). 9. Keogh, M.-C. The Saccharomyces cerevisiae histone H2A variant Htz1 is acetylated by NuA4. Genes Dev. 20, 660–665 (2006). 10. Millar, C.B., Xu, F., Zhang, K. & Grunstein, M. Acetylation of H2AZ lysine 14 is associated with genome-wide gene activity in yeast. Genes Dev. 20, 711–722 (2006). 11. Babiarz, J.E., Halley, J.E. & Rine, J. Telomeric heterochromatin boundaries require NuA4-dependent acetylation of histone variant H2A.Z in Saccharomyces cerevisiae. Genes Dev. 20, 700–710 (2006). 12. Ren, Q. & Gorovsky, M.A. Histone H2A.Z acetylation modulates an essential charge patch. Mol. Cell 7, 1329–1335 (2001). 13. Bruce, K. et al. The replacement histone H2A.Z in a hyperacetylated form is a feature of active genes in the chicken. Nucleic Acids Res. 33, 5633–5639 (2005). 14. Bonenfant, D., Coulot, M., Towbin, H., Schindler, P. & van Oostrum, J. Characterization of histone H2A and H2B variants and their post-translational modifications by mass spectrometry. Mol. Cell. Proteomics 5, 541–552 (2006). 15. Krogan, N.J. et al. Regulation of chromosome stability by the histone H2A variant Htz1, the Swr1 chromatin remodeling complex and the histone acetyltransferase NuA4. Proc. Natl. Acad. Sci. USA 101, 13513–13518 (2004). 16. Gómez, E.B., Nugent, R.L., Laria, S. & Forsburg, S.L. S. pombe histone acetyltransferase Mst1 (KAT5) is an essential protein required for damage response and chromosome segregation. Genetics 179, 757–771 (2008). 17. Krogan, N.J. et al. A Snf2-Family ATPase complex required for recruitment of the histone H2A variant Htz1. Mol. Cell 12, 1565–1576 (2003). 18. Kobor, M.S. et al. A protein complex containing the conserved Swi2/Snf2-related ATPase Swr1p deposits histone variant H2A.Z into euchromatin. PLoS Biol. 2, e131 (2004). 19. Mizuguchi, G. et al. ATP-driven exchange of histone H2AZ variant catalyzed by SWR1 chromatin remodeling complex. Science 303, 343–348 (2004). 20. Roguev, A. et al. Conservation and rewiring of functional modules revealed by an epistasis map (E-MAP) in fission yeast. Science 322, 405–410 (2008). 21. Roguev, A., Wiren, M., Weissman, J.S. & Krogan, N.J. High-throughput genetic interaction mapping in the fission yeast Schizosaccharomyces pombe. Nat. Methods 4, 861–866 (2007). 22. Schuldiner, M. et al. Exploration of the function and organization of the yeast early secretory pathway through an epistatic miniarray profile. Cell 123, 507–519 (2005). 23. Collins, S.R. et al. Functional dissection of protein complexes involved in yeast chromosome biology using a genetic interaction map. Nature 446, 806–810 (2007). 24. Collins, S.R., Schuldiner, M., Krogan, N.J. & Weissman, J.S. A strategy for extracting and analyzing large-scale quantitative epistatic interaction data. Genome Biol. 7, R63 (2006). 25. Carr, A.M. et al. Analysis of a histone H2A variant from fission yeast: evidence for a role in chromosome stability. Mol. Gen. Genet. 245, 628–635 (1994). 26. Ahmed, S., Dul, B., Qiu, X. & Walworth, N.C. Msc1 acts through histone H2A.Z to promote chromosome stability in Schizosaccharomyces pombe. Genetics 177, 1487–1497 (2008). 27. Ekwall, K. Epigenetic control of centromere behaviour. Annu. Rev. Genet. 41, 63–81 (2007). 28. Gregan, J. et al. The kinetochore proteins Pcs1 and Mde4 and heterochromatin are required to prevent merotelic orientation. Curr. Biol. 17, 1190–1200 (2007). 29. Cooper, J.P., Nimmo, E.R., Allshire, R.C. & Cech, T.R. Regulation of telomere length and function by a Myb-domain protein in fission yeast. Nature 385, 744–747 (1997).

30. Ding, D.-Q., Yamamoto, A., Haraguchi, T. & Hiraoka, Y. Dynamics of homologous chromosome pairing during meiotic prophase in fission yeast. Dev. Cell 6, 329–341 (2004). 31. Vanoosthuyse, V., Prykhozhij, S. & Hardwick, K.G. Shugoshin 2 regulates localization of the chromosomal passenger proteins in fission yeast mitosis. Mol. Biol. Cell 18, 1657–1669 (2007). 32. Kawashima, S.A. et al. Shugoshin enables tension-generating attachment of kinetochores by loading Aurora to centromeres. Genes Dev. 21, 420–435 (2007). 33. Nakazawa, N. et al. Dissection of the essential steps for condensin accumulation at kinetochores and rDNAs during fission yeast mitosis. J. Cell Biol. 180, 1115–1131 (2008). 34. Saka, Y. et al. Fission yeast cut3 and cut14, members of a ubiquitous protein family, are required for chromosome condensation and segregation in mitosis. EMBO J. 13, 4938–4952 (1994). 35. Sutani, T. et al. Fission yeast condensin complex: essential roles of non-SMC subunits for condensation and Cdc2 phosphorylation of Cut3/SMC4. Genes Dev. 13, 2271–2283 (1999). 36. Hagstrom, K.A., Holmes, V.F., Cozzarelli, N.R. & Meyer, B.J. C. elegans condensin promotes mitotic chromosome architecture, centromere organization, and sister chromatid segregation during mitosis and meiosis. Genes Dev. 16, 729–742 (2002). 37. Hiraoka, Y., Toda, T. & Yanagida, M. The NDA3 gene of fission yeast encodes β-tubulin: a cold-sensitive nda3 mutation reversibly blocks spindle formation and chromosome movement in mitosis. Cell 39, 349–358 (1984). 38. Chen, E.S. et al. Cell cycle control of centromeric repeat transcription and heterochromatin assembly. Nature 451, 734–737 (2008). 39. Sipiczki, M. Where does fission yeast sit on the tree of life? Genome Biol. 1, 1011 (2000). 40. O’Brien, S.J. et al. The promise of comparative genomics in mammals. Science 286, 458–462 (1999). 41. Takahata, N. & Satta, Y. Evolution of the primate lineage leading to modern humans: phylogenetic and demographic inferences from DNA sequences. Proc. Natl. Acad. Sci. USA 94, 4811–4815 (1997). 42. Shevchenko, A. et al. Chromatin central: towards the comparative proteome by accurate mapping of the yeast proteomic environment. Genome Biol. 9, R167 (2008). 43. Ahmed, S., Palermo, C., Wan, S. & Walworth, N.C. A novel protein with similarities to Rb binding protein 2 compensates for loss of chk1 function and affects histone modification in fission yeast. Mol. Cell. Biol. 24, 3660–3669 (2004). 44. Choi, K. et al. Arabidopsis homologs of components of the SWR1 complex regulate flowering and plant development. Development 134, 1931–1941 (2007). 45. Ruhl, D.D. et al. Purification of a human SCRAP complex that remodels chromatin by incorporating the histone variant H2A.Z into nucleosomes. Biochemistry 45, 5671–5677 (2006). 46. Kusch, T. et al. Acetylation by Tip60 is required for selective histone variant exchange at DNA lesions. Science 306, 2084–2087 (2004). 47. Updike, D.L. & Mango, S.E. Temporal regulation of foregut development by HTZ-1/H2A.Z and PHA-4/FoxA. PLoS Genet. 2, e161 (2006). 48. Vagnarelli, P. et al. Condensin and Repo-Man-PP1 co-operate in the regulation of chromosome architecture during mitosis. Nat. Cell Biol. 8, 1133–1142 (2006). 49. Fiedler, D. et al. Functional organization of the S. cerevisiae phosphorylation network. Cell 136, 952–963 (2009). 50. Nabetani, A., Koujin, T., Tsutsumi, C., Haraguchi, T. & Hiraoka, Y. A conserved protein, Nuf2, is implicated in connecting the centromere to the spindle during chromosome segregation: a link between the kinetochore function and the spindle checkpoint. Chromosoma 110, 322–334 (2001). 51. Gulli, M.P. et al. gar2 is a nucleolar protein from Schizosaccharomyces pombe required for 18S rRNA and 40S ribosomal subunit accumulation. Nucleic Acids Res. 23, 1912–1918 (1995). 52. Hagstrom, K.A. & Meyer, B.J. Condensin and cohesin: more than chromosome compactor and glue. Nat. Rev. Genet. 4, 520–534 (2003). 53. Hirano, T. Condensins: Organizing and segregating the genome. Curr. Biol. 15, R265–R275 (2005).

nature structural & molecular biology  VOLUME 16  NUMBER 12  december 2009

1293

ONLINE METHODS Materials. Antibodies, strains and oligos are listed in Supplementary Tables 3–5, respectively. Peroxidase anti-peroxidase (antiPAP) to recognize the TAP tag in immunoblotting was obtained from Sigma. Rabbit polyclonal ­affinity-purified anti-Sp H2A.Z antibodies were from Active Motif. C-terminal anti-Pht1 (no. 39640) was raised against residues 121–134 (C-KQLLIRTKEKYPEE), anti-Pht1Ac (no. 39642) against residues 1–20 (SGGG[KAc]G[KAc]HVGG [KAc]GGS[KAc]IGER-C) (Fig. 1a). We used the terminal C residue on each peptide to couple to keyhole limpet hemocyanin for immunization or a sulfolink resin for affinity purification.

© 2009 Nature America, Inc. All rights reserved.

Cell fractionation. Sp cell fractionations were performed essentially as described for Sc9, with cultures harvested at OD600 ~0.5. Total, cytoplasmic, nuclear and chromatin fractions were analyzed by SDS-PAGE and immunoblotting. Cell-cycle synchronization: ChIP and septation analyses. Cell-cycle synchronization by cdc25-22 was as previously described33,54. Briefly, cultures were G2-arrested at the nonpermissive temperature (36.5 °C, 4.5 h) before synchronized release at 24.5 °C. Aliquots were collected at 25-min intervals and fixed either with formaldehyde for ChIP (50 ml) or in 70% (v/v) ethanol for cell-­septation analysis (500 µl). The latter were collected by flash spin centrifugation at 10,000 rpm (Eppendorf 5810R with microfuge rotor F45-30-11), washed in PBS, and resuspended in PBS + Calcofluor (25 µg ml–1). Septum formation was determined by microscopy under UV excitation with >300 cells counted per time point. Gene expression microarrays. Total RNA was isolated using a hot-phenol protocol55. 10–20 µg of total RNA were labeled by direct incorporation of either Cy3- or Cy5-dCTP (GE Healthcare), and the fluorescently labeled product was hybridized to Sp cDNA microarrays55. Microarrays were scanned with a GenePix 4000B laser scanner (Axon Instruments), fluorescence-intensity ratios were calculated with GenePix Pro (Axon Instruments) and data were normalized using a previously described script55. At least three biological repeats were analyzed for each mutant with dye swaps. To analyze the data, repeats for every given mutant were averaged. The significance of overlaps between different gene lists was calculated in GeneSpring (Agilent) using a standard Fisher’s exact test, and P values adjusted with a Bonferroni multiple testing correction. Microscopic analysis of mitotic chromosomes. Sp cells with green fluorescent protein (GFP)-tagged factors were grown in rich medium and briefly (< 30 s) fixed in 100% methanol before observation. DNA was visualized by DAPI staining. Imaging was performed with an Intelligent Imaging Innovations (3i) Marianas system. This uses a Zeiss Axiovert fluorescence microscope, a CoolSNAP HQ charge-coupled device camera (Photometrics) and Slidebook software (3i; Photometrics). Mini-chromosome loss assay. Deletions were mated into a Ch16 mini-chromosome strain containing episomal ade6-M216 to complement genomic ade6-M210 in trans56. The presence of both alleles results in white colonies; loss of Ch16 leads to red. Strains from a white colony were patched overnight onto YES, resuspended and dispensed onto plates with limiting adenine to allow optimal color development. Red, white and sectored colonies were counted and chromosome loss rates calculated. PEM2 analysis. Genetic screening of pht1 mutants was performed with the PEM-2 system20,21. NAT-marked mutant queries in the h– PEM2 background (p392 (KFP171); Supplementary Table 4) were mated against the Bioneer G418­resistant deletion set (h+, ura4-D18, leu1-32, ade6-M21X, yfg∆KanMX). Mating, haploid selection, data acquisition and analysis were as previously described20. Pair correlation coefficients (CCs) to determine the relationship between genetic screens (as in Figs. 4b,c) were calculated by the CORREL function (Excel). Telomere length by Southern blotting. Genomic DNA was isolated by standard glass bead phenol extraction. 20 µg of genomic DNA was digested with EcoRI, resolved on a 1% (w/v) agarose gel, transferred to a

nature structural & molecular biology

Hybond N+ membrane and probed with radiolabeled 500-bp synthetic telomeric repeats (gift from J. Cooper, Cancer Research UK). TAP purification of protein complexes. Briefly, 8 l of each C-terminally TAPtagged strain was grown at 30 °C to OD600 ~2.5 in YES medium. Cells were washed twice with distilled, deionized water, resuspended in 0.25 volume buffer E (20 mM HEPES, pH 8.0, 350 mM NaCl, 0.1% Tween-20 plus protease inhibitors) and frozen as noodles in liquid nitrogen. Frozen cells were mixed with dry ice, reduced to powder in a coffee grinder, adjusted to 50 ml total volume with buffer E, and lysed using a Biospec BeadBeater homogenizer lysed by agitation with an equal volume of glass beads (7 × 20 s pulses with 3 minutes on ice between bursts). Extracts were clarified by ultracentrifugation in a 70Ti rotor (30 min, 4 °C, 90,000g). Supernatants were collected in 50-ml Falcon tubes and directly added to 200 µl IgG-Sepharose (GE Healthcare) equilibrated in buffer E. Sequential affinity purification on IgG-Sepharose and calmodulin-Sepharose (GE Healthcare) was as before57. Half of the affinity-purified protein was vacuum concentrated, resolved by SDS-PAGE and silver stained, with gel slices trypsinized and analyzed by MALDI-TOF. The remainder of the pool was analyzed directly by LC-MS as below. LC-MS. Protein complexes were digested at 37 °C overnight in 50 mM NH4HCO3 (pH 8.5, 1 mM CaCl2) with a 1:30 molar ratio of recombinant proteomics-grade trypsin (Roche Diagnostics). The resulting peptide mixtures were dried with a SpeedVac (Savant) and reconstituted in 30 µl buffer A (5% v/v acetonitrile, 0.1% v/v formic acid). Peptides were directly loaded from a 96-well microplate autosampler using the EASY-nLC system (Proxeon Biosystems) at 3 µl min–1 to a 3 cm precolumn (150 µm i.d.) containing a Kasil frit packed with 5µ Magic C18 100 Å reversed-phase material (Michrom Bioresources). The precolumn was connected via a titanium micro-tee splitter fitted with an electrode for voltage application (2.3 kV) in a vented column set-up to an 8-cm fused silica microcapillary analytical column (75 µm i.d.) with a homemade laser-pulled spray tip packed with 5µ Magic C18 100 Å reversed-phase resin. For the peptide separation on the analytical column, a water-acetonitrile gradient was applied at an effective flow rate of 400 nl min–1, controlled by the EASY-nLC using a 2-h gradient. All samples were analyzed on an LTQ-Orbitrap XL. The instrument method consisted of one MS full scan (400–1,800 m/z) in the Orbitrap mass analyzer, an automatic gain control (AGC) target of 500,000 with a maximum ion injection of 500 ms, one microscan at a resolution of 60,000, and use of the preview scan option. Six data-dependent MS/MS scans were performed in the linear ion trap using the three most intense ions at 35% normalized collision energy. The MS and MS/MS scans were obtained in parallel. AGC targets were 10,000 with a maximum ion injection time of 100 ms. A minimum ion intensity of 1,000 was required to trigger an MS/MS spectrum. The dynamic exclusion was applied using a maximum exclusion list of 500 with one repeat count with a repeat duration of 30 s and exclusion duration of 45 s. Protein identification and data analysis. Raw files were converted to m/zXML using ReAdW and searched by X!Tandem against an Sp database containing known contaminants (human keratins and trypsin). Search parameters were as follows: fragment ion mass tolerance of 0.40 Da and a parent ion tolerance of ±10 ppm. Complete tryptic digestion was assumed. No fixed modifications were specified and oxidation of methionine was specified as a variable modification. Only proteins identified with two or more unique peptides and a protein expectation score (loge ≤ –3) were considered a positive identification, virtually eliminating false positive identifications in the reported dataset. 54. Dunaway, S. & Walworth, N.C. Assaying the DNA damage checkpoint in fission yeast. Methods 33, 260–263 (2004). 55. Lyne, R. et al. Whole-genome microarrays of fission yeast: characteristics, accuracy, reproducibility, and processing of array data. BMC Genomics 4, 27 (2003). 56. Niwa, O., Matsumoto, T. & Yanagida, M. Construction of a mini-chromosome by deletion and its mitotic and meiotic behaviour in fission yeast. Mol. Genet. Genomics 203, 397–405 (1986). 57. Krogan, N.J. et al. RNA polymerase II elongation factors of Saccharomyces cerevisiae: a targeted proteomics approach. Mol. Cell. Biol. 22, 6979–6992 (2002).

doi:10.1038/nsmb.1688

An acetylated form of histone H2A.Z regulates ...

Nov 15, 2009 - phenotypes are shared by an unacetylatable htz1 mutant, suggesting ... Received 5 January; accepted 10 September; published online 15 ..... Red dots indicate the CC of each genetic screen to the mutants on the x or y axis.

2MB Sizes 6 Downloads 146 Views

Recommend Documents

Cardiac hypertrophy and histone deacetylase ...
microliters lysine developer was added, incubated for. 30 minutes, and the absorbance at 405 ..... Data are shown as mean ± SEM. AP < 0.05 versus baseline.

Cotranscriptional Set2 Methylation of Histone H3 ...
Nov 17, 2005 - University of North Carolina School of Medicine. Chapel Hill, North Carolina 27599. 4 Howard Hughes ..... interactions and cluster together.

Combined targeting of histone deacetylases and ... -
pathways, activation of oncogenes and silencing of tumor suppressor genes.3 .... deregulation of gene transcription involved in controlling cell cycle progression ...

Cardiac hypertrophy and histone deacetylase ...
Phone: (215) 898-8731;. Fax: (215) .... and 6-week-old Hop transgenic mice (n = 6) were com- .... trichrome staining of myocardium from wild-type 28-week-old.

Combined targeting of histone deacetylases and ... -
47 +/- 2. 0.0001*. SAHA 5 µM + SANT-1 50 µM. 37 +/- 6. 0.004*. 46 +/- 3. 0.0001*. SAHA 5 µM + ..... Ongoing studies aim to evaluate the in vivo efficacy of .... mesh filter (Falcon) and analyzed by fluorescence activated cell sorting (FACS) on a .

Cardiac hypertrophy and histone deacetylase–dependent ... - JCI
Jul 15, 2003 - bars) mice between 5 and 10 weeks of age are shown. Hop trans- genic hearts are significantly enlarged. (c) M-mode echocardiogra- phy demonstrates thickened myocardial walls in transgenic mice (red arrows) and hyperdynamic function wit

Activation of a meiotic checkpoint regulates translation ...
Bentley, N. J. & Carr, A. M. DNA structure-dependent checkpoints in model systems. Biol. Chem. 378, 1267–1274 (1997). 21. Lasko, P. F. & Ashburner, M. The product of the Drosophila gene vasa is very similar to eukaryotic initiation factor-4A. Natur

Neuronal activity regulates the developmental ...
Available online on ScienceDirect (www.sciencedirect.com). ..... Multi-promoter system of rat BDNF .... data provide the additional information that deprivation of visual ..... Egan, M.F., Kojima, M., Callicott, J.H., Goldberg, T.E., Kolachana, B.S.,

Mammalian histone acetyltransferases and their ... - Springer Link
These findings provide us with necessary tools to address further questions of the precise ..... Consistent with this interpretation, comparison of single- and ..... 81 Dikstein R., Ruppert S. and Tjian R. (1996) TAFII250 is a bi- partite protein kin

Spatial organization of the extracellular matrix regulates ...
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. ...... Weber GF, Bjerke MA, DeSimone DW (2011) Integrins and cadherins join .... Hoechst in 10× video-microscopy acquisition and a 15-min time frame.

An Application of Bayesian Networks in Predicting Form ...
commerce and e-service domains where filling out infor- mation is strongly required. For instance, when we have to buy a book or a plane ticket, application may require to fill out many ... over the Internet. A recent ... business (B2B) aspects [1].

MODEL FORM OF DECLARATION OF INCOME IN THE FORM OF ...
MODEL FORM OF DECLARATION OF INCOME IN THE FORM OF AFFIDAVIT.PDF. MODEL FORM OF DECLARATION OF INCOME IN THE FORM OF ...

CIN85 regulates dopamine receptor endocytosis and ...
The EMBO Journal advance online publication, 15 June 2010; ..... course. To confirm the involvement of CIN85 in D2DR inter- nalization, we additionally performed a receptor-ligand-bind- ing assay, in which the binding of [3H]spiperone, a tritium- ...

REST Regulates Distinct Transcriptional Networks in ...
Oct 28, 2008 - use, distribution, and reproduction in any medium, provided the original author and source are credited. ...... emerged as tractable and meaningful in vitro models in which to use ..... (GE Healthcare). Non-IP DNA (Input, 250 ...

Glucose regulates preproinsulin messenger RNA levels ...
Data, determined by densitometry of Northern blots, are representative of ... recovery period in culture of up to 24 h [17,20]. .... Academic Press, New York.

Prion protein (PrP ) positively regulates neural ...
Feb 28, 2006 - in vitro'' (1 DIV)], 75% of WT cells express nestin (Fig. 3 A and. A ). Over the ..... were used: PrPc (goat polyclonal, 1:500; Abcam, Inc., Cambridge, .... Zhang, C. C., Steele, A. D., Lindquist, S. L. & Lodish, H. F. (2006) Proc. Nat

High Current Step-Down Controller Regulates to ... - Linear Technology
a DC/DC converter solution with high efficiency and small size. The LTC3832 also ... soft-start voltage ramp rate in this circuit is set by CSS. Figure 4 shows the ...

Prion protein (PrP ) positively regulates neural ...
Feb 28, 2006 - 2F) or O4-positive oligodendroglia (data not shown). ... †A.D.S. and J.G.E. contributed equally to this work. §S.L. and J.D.M. ... To whom correspondence may be addressed at: MGH-HMS Center for Nervous System. Repair ...

Serum ferritin regulates blood vessel formation: A ... - Semantic Scholar
Feb 10, 2009 - (1995) Role for NF-κB in the regulation of ferritin H by tumor necrosis factor-α. J Biol Chem 270:15285–15293. 11. Chen TT, et al. (2005) TIM-2 is expressed on B cells and in liver and kidney and is a receptor for H-ferritin endocy

ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice ...
4Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire. ...... tion of a constitutively active, nuclear-targeted MEK-ERK fusion construct ...

REREa/Atrophin-2 interacts with histone deacetylase ...
Jun 20, 2007 - ataxias (SCA 1, 2, 3, 6, and 7). .... plied (data not shown) and expressed .... 3. Disruption of REREa or histone deacety- lase (HDAC) activity causes a ...... from the Tübingen stock center for ... tier Science Program, and work in.

Membership Form of ICRI14 - MSBIRIA
PLEASE ATTACH ONE PAGE BIO-DATA WITH THIS FORM. DECLARATION. I, (full name) ... or amended from time to time. Date:______ Signature of Applicant: ...

WSTF regulates the H2A.X DNA damage response ... - Semantic Scholar
Dec 17, 2008 - response and expands our knowledge of domains that contain intrinsic ..... Full Methods and any associated references are available in the ...

Weight loss regulates inflammation-related genes in ...
file of obese subjects through a decrease of proin- flammatory factors and .... open-circuit ventilated canopy system (Deltatrac II monitor,. Datex Instrumentarium ...