Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Epigenetically induced paucity of histone H2A.Z stabilizes fission-yeast ectopic centromeres

Subjects

Abstract

In most eukaryotes, centromeres are epigenetically defined by nucleosomes that contain the histone H3 variant centromere protein A (CENP-A). Specific targeting of the CENP-A–loading chaperone to the centromere is vital for stable centromere propagation; however, the existence of ectopic centromeres (neocentromeres) indicates that this chaperone can function in different chromatin environments. The mechanism responsible for accommodating the CENP-A chaperone at noncentromeric regions is poorly understood. Here, we report the identification of transient, immature neocentromeres in Schizosaccharomyces pombe that show reduced association with the CENP-A chaperone Scm3, owing to persistence of the histone H2A variant H2A.Z. After the acquisition of adjacent heterochromatin or relocation of the immature neocentromeres to subtelomeric regions, H2A.Z was depleted and Scm3 was replenished, thus leading to subsequent stabilization of the neocentromeres. These findings provide new insights into histone variant–mediated epigenetic control of neocentromere establishment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Identification and characterization of Δcen3-NCs.
Figure 2: Reduced association of Cnp1 and Scm3 with Δcen3-NCs.
Figure 3: Defective chromosome III segregation in Δcen3-NC survivors.
Figure 4: Emergence of type-F and type-R revertants from Δcen3-NC cells.
Figure 5: Pht1 depletion promotes Cnp1 enrichment through efficient Scm3 recruitment.
Figure 6: Loss of rDNA repeats permits efficient NC formation on chromosome III.
Figure 7: The contribution of heterochromatin to Δcen3-NC maturation.
Figure 8: Schematic model summarizing ectopic centromere establishment in S. pombe.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Allshire, R.C. & Karpen, G.H. Epigenetic regulation of centromeric chromatin: old dogs, new tricks? Nat. Rev. Genet. 9, 923–937 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Santaguida, S. & Musacchio, A. The life and miracles of kinetochores. EMBO J. 28, 2511–2531 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Watanabe, Y. Geometry and force behind kinetochore orientation: lessons from meiosis. Nat. Rev. Mol. Cell Biol. 13, 370–382 (2012).

    Article  CAS  PubMed  Google Scholar 

  4. Marshall, O.J., Chueh, A.C., Wong, L.H. & Choo, K.H. Neocentromeres: new insights into centromere structure, disease development, and karyotype evolution. Am. J. Hum. Genet. 82, 261–282 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Burrack, L.S. & Berman, J. Neocentromeres and epigenetically inherited features of centromeres. Chromosome Res. 20, 607–619 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Van Hooser, A.A. et al. Specification of kinetochore-forming chromatin by the histone H3 variant CENP-A. J. Cell Sci. 114, 3529–3542 (2001).

    CAS  PubMed  Google Scholar 

  7. Choi, E.S. et al. Factors that promote H3 chromatin integrity during transcription prevent promiscuous deposition of CENP-A(Cnp1) in fission yeast. PLoS Genet. 8, e1002985 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ishii, K. et al. Heterochromatin integrity affects chromosome reorganization after centromere dysfunction. Science 321, 1088–1091 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Folco, H.D., Pidoux, A.L., Urano, T. & Allshire, R.C. Heterochromatin and RNAi are required to establish CENP-A chromatin at centromeres. Science 319, 94–97 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kagansky, A. et al. Synthetic heterochromatin bypasses RNAi and centromeric repeats to establish functional centromeres. Science 324, 1716–1719 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Olszak, A.M. et al. Heterochromatin boundaries are hotspots for de novo kinetochore formation. Nat. Cell Biol. 13, 799–808 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Alonso, A., Hasson, D., Cheung, F. & Warburton, P.E. A paucity of heterochromatin at functional human neocentromeres. Epigenetics Chromatin 3, 6 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Shang, W.H. et al. Chromosome engineering allows the efficient isolation of vertebrate neocentromeres. Dev. Cell 24, 635–648 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ketel, C. et al. Neocentromeres form efficiently at multiple possible loci in Candida albicans. PLoS Genet. 5, e1000400 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Yuen, K.W., Nabeshima, K., Oegema, K. & Desai, A. Rapid de novo centromere formation occurs independently of heterochromatin protein 1 in C. elegans embryos. Curr. Biol. 21, 1800–1807 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dunleavy, E.M. et al. HJURP is a cell-cycle-dependent maintenance and deposition factor of CENP-A at centromeres. Cell 137, 485–497 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Foltz, D.R. et al. Centromere-specific assembly of CENP-a nucleosomes is mediated by HJURP. Cell 137, 472–484 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Stoler, S. et al. Scm3, an essential Saccharomyces cerevisiae centromere protein required for G2/M progression and Cse4 localization. Proc. Natl. Acad. Sci. USA 104, 10571–10576 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mizuguchi, G., Xiao, H., Wisniewski, J., Smith, M.M. & Wu, C. Nonhistone Scm3 and histones CenH3–H4 assemble the core of centromere-specific nucleosomes. Cell 129, 1153–1164 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Camahort, R. et al. Scm3 is essential to recruit the histone H3 variant Cse4 to centromeres and to maintain a functional kinetochore. Mol. Cell 26, 853–865 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Williams, J.S., Hayashi, T., Yanagida, M. & Russell, P. Fission yeast Scm3 mediates stable assembly of Cnp1/CENP-A into centromeric chromatin. Mol. Cell 33, 287–298 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pidoux, A.L. et al. Fission yeast Scm3: a CENP-A receptor required for integrity of subkinetochore chromatin. Mol. Cell 33, 299–311 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jansen, L.E., Black, B.E., Foltz, D.R. & Cleveland, D.W. Propagation of centromeric chromatin requires exit from mitosis. J. Cell Biol. 176, 795–805 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Fujita, Y. et al. Priming of centromere for CENP-A recruitment by human hMis18α, hMis18β, and M18BP1. Dev. Cell 12, 17–30 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Silva, M.C. et al. Cdk activity couples epigenetic centromere inheritance to cell cycle progression. Dev. Cell 22, 52–63 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Hayashi, T. et al. Mis16 and Mis18 are required for CENP-A loading and histone deacetylation at centromeres. Cell 118, 715–729 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Pidoux, A.L., Richardson, W. & Allshire, R.C. Sim4: a novel fission yeast kinetochore protein required for centromeric silencing and chromosome segregation. J. Cell Biol. 161, 295–307 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Saitoh, S., Takahashi, K. & Yanagida, M. Mis6, a fission yeast inner centromere protein, acts during G1/S and forms specialized chromatin required for equal segregation. Cell 90, 131–143 (1997).

    Article  CAS  PubMed  Google Scholar 

  29. 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).

    Article  CAS  PubMed  Google Scholar 

  30. Takahashi, K., Chen, E.S. & Yanagida, M. Requirement of Mis6 centromere connector for localizing a CENP-A-like protein in fission yeast. Science 288, 2215–2219 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Tanaka, K., Chang, H.L., Kagami, A. & Watanabe, Y. CENP-C functions as a scaffold for effectors with essential kinetochore functions in mitosis and meiosis. Dev. Cell 17, 334–343 (2009).

    Article  PubMed  Google Scholar 

  32. Goshima, G., Saitoh, S. & Yanagida, M. Proper metaphase spindle length is determined by centromere proteins Mis12 and Mis6 required for faithful chromosome segregation. Genes Dev. 13, 1664–1677 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Takahashi, K. et al. A low copy number central sequence with strict symmetry and unusual chromatin structure in fission yeast centromere. Mol. Biol. Cell 3, 819–835 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Funabiki, H., Hagan, I., Uzawa, S. & Yanagida, M. Cell cycle-dependent specific positioning and clustering of centromeres and telomeres in fission yeast. J. Cell Biol. 121, 961–976 (1993).

    Article  CAS  PubMed  Google Scholar 

  35. 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).

    Article  CAS  PubMed  Google Scholar 

  36. Niwa, O. & Yanagida, M. Triploid meiosis and aneuploidy in Schizosaccharomyces pombe: an unstable aneuploid disomic for chromosome III. Curr. Genet. 9, 463–470 (1985).

    Article  Google Scholar 

  37. Ekwall, K. et al. The chromodomain protein Swi6: a key component at fission yeast centromeres. Science 269, 1429–1431 (1995).

    Article  CAS  PubMed  Google Scholar 

  38. Ekwall, K. et al. Mutations in the fission yeast silencing factors clr4+ and rik1+ disrupt the localisation of the chromo domain protein Swi6p and impair centromere function. J. Cell Sci. 109, 2637–2648 (1996).

    CAS  PubMed  Google Scholar 

  39. Buchanan, L. et al. The Schizosaccharomyces pombe JmjC-protein, Msc1, prevents H2A.Z localization in centromeric and subtelomeric chromatin domains. PLoS Genet. 5, e1000726 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Zofall, M. et al. Histone H2A.Z cooperates with RNAi and heterochromatin factors to suppress antisense RNAs. Nature 461, 419–422 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hou, H. et al. Histone variant H2A.Z regulates centromere silencing and chromosome segregation in fission yeast. J. Biol. Chem. 285, 1909–1918 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Kim, H.S. et al. An acetylated form of histone H2A.Z regulates chromosome architecture in Schizosaccharomyces pombe. Nat. Struct. Mol. Biol. 16, 1286–1293 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Takayama, Y. et al. Biphasic incorporation of centromeric histone CENP-A in fission yeast. Mol. Biol. Cell 19, 682–690 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lando, D. et al. Quantitative single-molecule microscopy reveals that CENP-A(Cnp1) deposition occurs during G2 in fission yeast. Open Biol. 2, 120078 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Greaves, I.K., Rangasamy, D., Ridgway, P. & Tremethick, D.J. H2A.Z contributes to the unique 3D structure of the centromere. Proc. Natl. Acad. Sci. USA 104, 525–530 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Choi, E.S. et al. Identification of noncoding transcripts from within CENP-A chromatin at fission yeast centromeres. J. Biol. Chem. 286, 23600–23607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gascoigne, K.E. et al. Induced ectopic kinetochore assembly bypasses the requirement for CENP-A nucleosomes. Cell 145, 410–422 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hori, T., Shang, W.H., Takeuchi, K. & Fukagawa, T. The CCAN recruits CENP-A to the centromere and forms the structural core for kinetochore assembly. J. Cell Biol. 200, 45–60 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Hirano, T., Konoha, G., Toda, T. & Yanagida, M. Essential roles of the RNA polymerase I largest subunit and DNA topoisomerases in the formation of fission yeast nucleolus. J. Cell Biol. 108, 243–253 (1989).

    Article  CAS  PubMed  Google Scholar 

  51. Matsuzaki, H., Nakajima, R., Nishiyama, J., Araki, H. & Oshima, Y. Chromosome engineering in Saccharomyces cerevisiae by using a site-specific recombination system of a yeast plasmid. J. Bacteriol. 172, 610–618 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Krawchuk, M.D. & Wahls, W.P. High-efficiency gene targeting in Schizosaccharomyces pombe using a modular, PCR-based approach with long tracts of flanking homology. Yeast 15, 1419–1427 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Masuda, H., Fong, C.S., Ohtsuki, C., Haraguchi, T. & Hiraoka, Y. Spatiotemporal regulations of Wee1 at the G2/M transition. Mol. Biol. Cell 22, 555–569 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nabeshima, K. et al. Dynamics of centromeres during metaphase-anaphase transition in fission yeast: Dis1 is implicated in force balance in metaphase bipolar spindle. Mol. Biol. Cell 9, 3211–3225 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Yamamoto, A. & Hiraoka, Y. Monopolar spindle attachment of sister chromatids is ensured by two distinct mechanisms at the first meiotic division in fission yeast. EMBO J. 22, 2284–2296 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Toda, T., Nakaseko, Y., Niwa, O. & Yanagida, M. Mapping of rRNA genes by integration of hybrid plasmids in Schizosaccharomyces pombe. Curr. Genet. 8, 93–97 (1984).

    Article  CAS  PubMed  Google Scholar 

  57. Hagan, I.M. & Hyams, J.S. The use of cell division cycle mutants to investigate the control of microtubule distribution in the fission yeast Schizosaccharomyces pombe. J. Cell Sci. 89, 343–357 (1988).

    PubMed  Google Scholar 

  58. Woods, A. et al. Definition of individual components within the cytoskeleton of Trypanosoma brucei by a library of monoclonal antibodies. J. Cell Sci. 93, 491–500 (1989).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank Y. Hiraoka (Osaka University) for the GFP visualization plasmids, H. Matsuzaki (Fukuyama University) for the R-recombinase clone and K. Gull (University of Oxford) for the TAT1 antibodies. We also thank H. Kimura and T. Stasevich (Osaka University) for critical reading of the manuscript. This study was supported by Grants-in-Aid for Young Scientists (A) (K.I., 21687014), Scientific Research (B) (K.I., 24370003) and Challenging Exploratory Research (K.I., 22657002 and 25650122) from the Japan Society for the Promotion of Science (JSPS), Grant-in-Aid for Scientific Research on Innovative Areas (Genome Adaptation) (K.I., 22125004) from the Ministry of Education, Culture, Sports, Science and Technology of Japan, the Osaka University Life Science Young Independent Researcher Support Program (K.I.) and the Naito Foundation (K.I.). Y. Ogiyama and Y. Ohno are supported as JSPS Fellows.

Author information

Authors and Affiliations

Authors

Contributions

Y. Ogiyama and K.I. designed the experiments. Y. Ogiyama, Y.K. and K.I. performed the experiments. Y. Ogiyama and K.I. analyzed the data. Y. Ohno performed the sequencing analysis of NC-accommodating genomes. Y. Ogiyama and K.I. interpreted the data, and K.I. wrote the paper.

Corresponding author

Correspondence to Kojiro Ishii.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Analysis of Cnp1 association with NCs.

(a,b) The chromosomal distribution of Cnp1 (red) and subtelomeric heterochromatin (dimethylated histone H3 K9 (H3K9me2); blue) along the Δcen1-NCs (left subtelomeric region (tel1-L, a) and right subtelomeric region (tel1-R, b)) in the indicated strains. A repeat-suppressed S. pombe genomic microarray was used for the ChIP-chip analysis. The repetitive regions suppressed in the microarray are indicated in yellow. The x-axis numbers represent the chromosome I coordinates. The NCs in cd1-46 and cd1-50 were previously mapped to the same loci as those in cd1-39 and cd1-60, respectively8, and exhibit similar Cnp1 distributions. The variation of Cnp1 distribution along the NCs infrequently formed in the absence of heterochromatin (Δclr4 cd1-132 and Δclr4 cd1-136)8 was within the range of that seen in the wild-type. For reference, the positions of the quantitative chromosome I PCR probes (a–e) used elsewhere in this study are also indicated. (c,d) The chromosomal distribution of Cnp1 along the Δcen2-NCs (cd2-163 (c) and cd2-166 (d)). A repeat covering S. pombe genomic microarray was used for the ChIP-chip analyses. The corresponding chromosome II regions (tel2-L (c) and tel2-R (d)) are displayed in alignment with the Sanger Center S. pombe genome coordinates. Annotated genes are indicated by arrows or arrowheads and essential genes are displayed in pink. (e) NC mapping of 20 Δcen2-NC survivors showing Cnp1 ChIP enrichments in each strain. All of the Δcen2-NCs were mapped to either tel2-L or tel2-R and none were mapped to the mat locus. (f,g) The chromosomal distribution of Cnp1 along the NCs obtained in the Δcen3 experiments (cd3-389 (f) and cd3-385 (g)). The ChIP-chip analysis and data presentation were performed as described in c,d. Only four Δcen3-NC survivors were obtained in total, and the ChIP-chip results for the two other survivors (Y. Ogiyama and K.I., unpublished data) were almost identical to those for cd3-389. For reference, the positions of the chromosome III quantitative PCR probes (a–j) used elsewhere in this study are also indicated.

Supplementary Figure 2 Irregular chromosome III segregation in Δcen3-NC cells.

(a) Quantitative ChIP enrichment of Cnp1 protein across the Δcen3-NCssp2 region in cd3-385 and the Δcen3-NCrDNA region in cd3-389. The positions of the quantitative PCR probes (a–j) in chromosomes III are shown in Supplementary Figure 1. The ChIP enrichments were normalized to those of the cnt2 region in cen2 (cen2). The data are represented as the average + s.e.m. of n = 3 biological repeats. (b) A typical fluorescent microscopy image of GFP-Cnp1-visualized centromeres (green), Nuc1-mCherry-visualized rDNAs50 (magenta), and Hoechst 33342-stained DNAs (blue) of Δcen3-NC cells (cd3-385) harboring the nda3-KM311 cold sensitive mutation35. The cells were arrested in mitosis by incubation at 18°C for 10 h. Cells exhibiting more than two GFP-Cnp1 dots connected to an rDNA signal, in addition to two other GFP-Cnp1 dots corresponding to canonical cen1 and cen2, are indicated by arrowheads. The cell that had lost chromosome III (no rDNA signal) is indicated by an arrow. Scale bar, 10 μm.(c) The percentages of cells exhibiting loss or gain of chromosome III. The indicated strains were mitotically arrested as described in b. The rDNA signals tended to be clustered and were difficult to distinguish even in the nda3-arrested nucleus; therefore, the numbers of GFP-Cnp1 dots associated with the rDNA-containing chromosome in each nucleus were counted. Increases in irregular numbers of rDNA-connected GFP-Cnp1 dots (colored bars) in Δcen3-NCrDNA cells (cd3-389) and Δcen3-NCssp2 cells (cd3-385) are indicative of chromosome III mis-segregation. 7.4% of the Δcen3-NCssp2 cells harbored an rDNA-containing chromosome III without any GFP-Cnp1 dots (cyan), suggesting a loss of NC from the chromosome. Note that the quantification of GFP-Cnp1 signal intensities shown in Figure 2d was performed using only the cells exhibiting a regular single GFP-Cnp1 dot connected to an rDNA signal (gray).

Supplementary Figure 3 Profiles of the centromere deletion screens and resultant survivor characterizations.

(a) The viabilities of total cells (blue) and centromere-deleted cells (magenta), as determined by their resistance to G418 and 5-fluoroorotic acid (see METHODS). Error bars represents s.e.m. of n = 6 biological replicates. loxP-cen1, loxP-cen2, and loxP-cen3, the strains used for deleting cen1, cen2, and cen3, respectively. (b) The relative ratios between NC formation and telomere fusion in the survivors obtained in each screen. (c) Cell growth of the wild-type, NC survivors, and revertant cells growing on YES media at 33°C. The generation times of each strain were as follows: wild-type, 2.1 h; cd1-39, 1.9 h; cd1-60, 2.0 h; cd2-163, 2.1 h; cd2-166, 2.2 h; cd3-385, 2.7 h; cd3-386, 3.0 h; and cd3-386-r1, 2.0 h. (d) A typical fluorescent microscopic image of Hoechst 33342-stained anaphase chromosomes (magenta). Chromosome III visualization was obtained by tandem tethering of the GFP-LacI protein (green). Chromosome III was segregated equally to the daughter nuclei in wild-type cells but not Δcen3-NC cells (cd3-385). Scale bar, 10 μm. (e) The frequency of chromosome I segregation failure in the defective cells displaying lagging chromosomes of the indicated strains (n > 50). Cells were cultured at 33°C. One locus in chromosome I was visualized by tandem tethering of the GFP-LacI protein. Error bars represents s.e.m. of triplicate cultures. One third of the lagging chromosomes observed in heterochromatin-abrogated Δclr4 cells were derived from chromosome I, irrespective of whether it harbored canonical cen1 or Δcen1-NC, suggesting that not every NC results in lagging chromosomes.

Supplementary Figure 4 The behavior of spontaneous revertants of Δcen3-NCs.

(a) A color image of a typical colony showing the emergence of spontaneous revertants from the Δcen3-NC strain. Δcen3-NC cells (cd3-386) were grown continuously for 30 generations and were allowed to form colonies on YES plates containing phloxine B (PhB). (b) The proportions of revertants that emerged from the indicated Δcen3-NC strains during continuous cell culture. (c) PFGE separation of the chromosomes in the type-F revertants showing the reduction in chromosome number. m, molecular size marker. (d) SfiI-digested chromosomes of the type-F revertants separated by PFGE and stained with ethidium bromide (EtBr) or subjected to Southern blot (SB) analysis with the chr3L, chr3R, tel1L, and tel2R probes to observe telomere-to-telomere fusion. (e) PFGE separation of the chromosomes in the type-R revertants showing the three-chromosome configurations. m, molecular size marker. (f) The percentages of normal (gray) and defective (magenta) chromosome segregation patterns in anaphase cells. The percentages were determined as described in Figure 3e (n > 100). Error bars represents s.e.m. of triplicate cultures. Note that segregation failure occurs less frequently in the type-R revertants than in the heterochromatin-deficient Δswi6 cells and Δclr4 cells (see Fig. 3e). Furthermore, the Δswi6 mutation in the type-R revertant conferred a low level of lagging chromosomes (cd3-389-r1 Δswi6). This level was similar to that observed in the Δswi6 mutant, but different from that observed in the original Δcen3-NCrDNA cells (cd3-389). (g) SfiI-digested chromosomes of the type-R revertants separated by PFGE and stained with EtBr or subjected to Southern blot analysis with the indicated probes to show the reduction in the number of rDNA repeats in the left arm. (h) PstI-digested chromosomes of the type-R revertants separated by PFGE and subjected to Southern blot analysis with the rDNA probe. Given that there is no PstI restriction site in the 10.8-kb rDNA repeat units and the non-rDNA repeat region of the left-most PstI fragment in chromosome III encompasses approximately 3.4 kb (chrIII, 24571–28056), the rDNA 15-kb PstI fragment in cd3-386-r1 and cd3-390-r1 that hybridized to the probe most likely harbors only one copy of the rDNA repeat, and the 25-kb PstI fragment in cd3-389-r1 most likely harbors two copies.

Supplementary Figure 5 Associations of Cnp1, Scm3, Pht1 and heterochromatin with Δcen3-NCs and derivatives.

(a,e) Quantification of Cnp1 association with Δcen3-NCrDNA (a) and Δcen3-NCssp2 (e). The ChIP enrichments were normalized to the cnt2 region in cen2 (cen2). The probe positions are indicated in Supplementary Figure 1. The data are represented as the average + s.e.m. of n = 3 biological repeats. (b,c,f,g) Changes in the association of Scm3-GFP (b,f) and Pht1-GFP (c,g) with Δcen3-NCs following the heterochromatin supplementation. The ChIP enrichments across the Δcen3-NCrDNA (b,c) and Δcen3-NCssp2 (f,g) regions were quantified and normalized to those of the cnt2 region in cen2 (cen2) (b,f), and the Pht1-enriched region (SPAC1486.08)40 (c,g), respectively. The probe positions (a–j) in chromosome III are shown in Supplementary Figure 1. Data are represented as the average + s.e.m. of n = 3 biological replicates. Like the type-R revertant (cd3-389-r1), the addition of heterochromatin to the Δcen3-NCrDNA region (cd3-389+dh2.1) caused Pht1 depletion, and the Δswi6 mutation37 reversed the Pht1 depletion effect (cd3-389+dh2.1 Δswi6) (c). A similar effect was observed, albeit to a lesser extent, at the Δcen3-NCssp2 region (g). The changes in Scm3 association with Δcen3-NCs were inversely proportional to the changes in Pht1 association (b,f). However, Scm3 accumulation in the type-R revertant was not reversed by the Δswi6 mutation (cd3-389-r1 Δswi6) (b). (d,h) Quantification of artificial heterochromatin around Δcen3-NCrDNA (d) and Δcen3-NCssp2 (h). The ChIP enrichments were normalized to the heterochromatin at the canonical centromeres (cen-otr). The probe positions are indicated in Supplementary Figures 8c,d. Artificial heterochromatin formation was more prominent at the Δcen3-NCrDNA region than the Δcen3-NCssp2 region, which paralleled the replenishment of Cnp1 at the Δcen3-NCs (a,e). The data are represented as the average + s.e.m. of n = 3 biological repeats.

Supplementary Figure 6 The inverse relationship between Cnp1 and Pht1 association with NCs.

(a,b,f,g) Chromosomal distributions of Pht1-GFP (light green) and Cnp1 (magenta) in Δcen1-NC cells (cd1-39 (a) and cd1-60 (b)) and Δcen3-NC cells (cd3-389 (f) and cd3-385 (g)). Normalization and smoothing of ChIP-chip data were performed as described in Figure 5a. The 120-kb segments used for a higher magnification shown in Figure 5b ((i) to (iv)) are also indicated by blue lines. The apparent Cnp1 enrichments at the right terminus of chromosome III in cd3-389 (f) most likely result from false mapping of Cnp1 signals at the left terminus, because the right signals corresponded only to genomic regions that are conserved between both termini. (c) A higher magnification of the Pht1 distributions across the 120-kb segments encompassing canonical centromeres and NC-competent subtelomeric regions in wild-type (light green) and Δmsc1 cells (dark green). ChIP-chip data were processed as described for (a,b,f,g), except that an 11-probe window was used instead of a 201-probe window for the smoothing. The positions corresponding to Cnp1-containing centric chromatin and heterochromatin are indicated by filled rectangles. (d) Quantification of GFP-Cnp1 signals in wild-type and Δmsc1 cells in interphase. The analysis was performed as described in Figure 5c. a.u., arbitrary units. (e) Expression of the GFP-tagged scm3+ gene in wild-type, Δmsc1, and Δswi6 cells, as determined by quantitative RT-PCR. Data were normalized to act1+ expression levels and are represented as the average ± s.d. of n = 3 technical repeats.

Supplementary Figure 7 Centromere deletion screens of the t(1Rter;3Lter) and Δmsc1 strains.

(a) The viabilities of total cells (blue) and centromere-deleted cells (magenta) in the course of centromere deletion screens (loxP-cen1 and loxP-cen3) of the indicated strains. Error bars represents s.e.m. of n = 6 biological replicates. (b) The ratios of NC formation to telomere fusion in the survivors obtained in each screen. (c) NC mapping of six Δcen3-NC survivors obtained from the t(1Rter;3Lter) loxP-cen3 strain showing the Cnp1 ChIP enrichment values for each strain. All of the Δcen3-NCs were mapped to the region corresponding to Δcen3-NCrDNA (rDNA(L)). (d) NC mapping of six Δcen1-NC survivors obtained from the t(1Rter;3Lter) loxP-cen1 strain. All of the Δcen1-NCs were mapped to the left subtelomeric region of chromosome I (tel1-L) and none were mapped to the right subtelomeric region (tel1-R), even though NCs are formed more preferentially at tel1-R than tel1-L in wild-type cells8.

Supplementary Figure 8 The contribution of heterochromatin to Δcen3-NC maturation.

(a) A ChIP-chip analysis showing the acquisition of robust heterochromatin (H3K9me2; blue) near Δcen3-NCrDNA (Cnp1; red) in the type-R revertant (cd-389-r1, top), but not in the original Δcen3-NC cells (cd3-389, bottom). The yellow arrows indicate rDNA repeats. (b) Verification of the size of the rDNA repeats by PFGE of SfiI-digested chromosomes and subsequent Southern blot analysis (SB) with the indicated probes. Both the left and right termini of chromosome III (3L and 3R, respectively) showed similar size variations among the mock and dh2.1 integrants, indicating no marked reductions in the number of rDNA repeats. (c,d) ChIP-chip analysis confirming the accumulation of dimethylated histone H3 K9 modification (H3K9me2; blue) at the heterochromatin artificially formed next to the Cnp1-assembled NCs (red) in the Δcen3-NCrDNA strain (cd3-389) (c) and the Δcen3-NCssp2 strain (cd3-385) (d) following insertion of the dh2.1 DNA fragment (vertical arrow, top panels), but not following a mock insertion (bottom panels). The blue-shaded area indicates the integrated plasmid DNA region and the yellow arrows indicate rDNA repeats.

Supplementary Figure 9 Original images of gels and blots corresponding to Figure 4c,e and a validation of antibodies.

(a) PFGE analysis. m, molecular size marker. (b) Southern blot analysis with the rDNA probe. (c) Western blot analysis of S. pombe whole cell lysates with anti-Swi6 antibodies.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1 and 2 (PDF 4363 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ogiyama, Y., Ohno, Y., Kubota, Y. et al. Epigenetically induced paucity of histone H2A.Z stabilizes fission-yeast ectopic centromeres. Nat Struct Mol Biol 20, 1397–1406 (2013). https://doi.org/10.1038/nsmb.2697

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2697

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing