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:

A two-step mechanism for epigenetic specification of centromere identity and function

Abstract

The basic determinant of chromosome inheritance, the centromere, is specified in many eukaryotes by an epigenetic mark. Using gene targeting in human cells and fission yeast, chromatin containing the centromere-specific histone H3 variant CENP-A is demonstrated to be the epigenetic mark that acts through a two-step mechanism to identify, maintain and propagate centromere function indefinitely. Initially, centromere position is replicated and maintained by chromatin assembled with the centromere-targeting domain (CATD) of CENP-A substituted into H3. Subsequently, nucleation of kinetochore assembly onto CATD-containing chromatin is shown to require either the amino- or carboxy-terminal tail of CENP-A for recruitment of inner kinetochore proteins, including stabilizing CENP-B binding to human centromeres or direct recruitment of CENP-C, respectively.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: CENP-A conditional disruption in human cells.
Figure 2: Disrupted centromere positioning and kinetochore nucleation requires almost complete loss of CENP-A.
Figure 3: Short-term rescue of centromere maintenance and function following CENP-A gene depletion.
Figure 4: H3CATD is sufficient for centromere identity and to template its chromatin replication.
Figure 5: N- or C-terminal tails of CENP-A nucleate kinetochore assembly.
Figure 6: The CENP-A N-terminal tail controls CENP-B levels at centromeres.
Figure 7: The fission yeast CATD is necessary and sufficient for centromere identity, but requires addition of the CENP-ACnp1 N terminus to provide long-term centromere function.
Figure 8: Model for centromeric identity, maintenance and function through a two-step mechanism.

Similar content being viewed by others

References

  1. Palmer, D. K., O’Day, K., Wener, M. H., Andrews, B. S. & Margolis, R. L. A 17-kD centromere protein (CENP-A) copurifies with nucleosome core particles and with histones. J. Cell Biol. 104, 805–815 (1987).

    Article  CAS  PubMed  Google Scholar 

  2. Earnshaw, W. C. & Rothfield, N. Identification of a family of human centromere proteins using autoimmune sera from patients with scleroderma. Chromosoma 91, 313–321 (1985).

    Article  CAS  Google Scholar 

  3. Cleveland, D. W., Mao, Y. & Sullivan, K. F. Centromeres and kinetochores: from epigenetics to mitotic checkpoint signaling. Cell 112, 407–421 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Karpen, G. H. & Allshire, R. C. The case for epigenetic effects on centromere identity and function. Trends Genet. 13, 489–496 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Ekwall, K. Epigenetic control of centromere behavior. Annu. Rev. Genet. 41, 63–81 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Du Sart, D. et al. A functional neo-centromere formed through activation of a latent human centromere and consisting of non-alpha-satellite DNA. Nat. Genet. 16, 144–153 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Ventura, M. et al. Recurrent sites for new centromere seeding. Genome Res. 14, 1696–1703 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Amor, D. J. et al. Human centromere repositioning ‘in progress’. Proc. Natl Acad. Sci. USA 101, 6542–6547 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Warburton, P. E. Chromosomal dynamics of human neocentromere formation. Chromosome Res. 12, 617–626 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Depinet, T. W. et al. Characterization of neo-centromeres in marker chromosomes lacking detectable alpha-satellite DNA. Hum. Mol. Genet. 6, 1195–1204 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. Warburton, P. E. et al. Immunolocalization of CENP-A suggests a distinct nucleosome structure at the inner kinetochore plate of active centromeres. Curr. Biol. 7, 901–904 (1997).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. Schuh, M., Lehner, C. F. & Heidmann, S. Incorporation of Drosophila CID/CENP-A and CENP-C into centromeres during early embryonic anaphase. Curr. Biol. 17, 237–243 (2007).

    Article  CAS  PubMed  Google Scholar 

  14. Mellone, B. G. et al. Assembly of Drosophila centromeric chromatin proteins during mitosis. PLoS Genet. 7, e1002068 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dimitriadis, E. K., Weber, C., Gill, R. K., Diekmann, S. & Dalal, Y. Tetrameric organization of vertebrate centromeric nucleosomes. Proc. Natl Acad. Sci. USA 107, 20317–20322 (2010).

    Article  PubMed  Google Scholar 

  16. Furuyama, T. & Henikoff, S. Centromeric nucleosomes induce positive DNA supercoils. Cell 138, 104–113 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Krassovsky, K., Henikoff, J. G. & Henikoff, S. Tripartite organization of centromeric chromatin in budding yeast. Proc. Natl Acad. Sci. USA 109, 243–248 (2012).

    Article  PubMed  Google Scholar 

  18. 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 

  19. Conde e Silva, N. et al. CENP-A-containing nucleosomes: easier disassembly versus exclusive centromeric localization. J. Mol. Biol. 370, 555–573 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Camahort, R. et al. Cse4 is part of an octameric nucleosome in budding yeast. Mol. Cell 35, 794–805 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sekulic, N., Bassett, E. A., Rogers, D. J. & Black, B. E. The structure of (CENP-A-H4)(2) reveals physical features that mark centromeres. Nature 467, 347–351 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tachiwana, H. et al. Crystal structure of the human centromeric nucleosome containing CENP-A. Nature 476, 232–235 (2011).

    Article  CAS  Google Scholar 

  23. Zhang, W., Colmenares, S. U. & Karpen, G. H. Assembly of Drosophila centromeric nucleosomes requires CID dimerization. Mol. Cell 45, 263–269 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Black, B. E. & Cleveland, D. W. Epigenetic centromere propagation and the nature of CENP-a nucleosomes. Cell 144, 471–479 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Regnier, V. et al. CENP-A is required for accurate chromosome segregation and sustained kinetochore association of BubR1. Mol. Cell Biol. 25, 3967–3981 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Liu, S. T., Rattner, J. B., Jablonski, S. A. & Yen, T. J. Mapping the assembly pathways that specify formation of the trilaminar kinetochore plates in human cells. J. Cell Biol. 175, 41–53 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Black, B. E. et al. Centromere identity maintained by nucleosomes assembled with histone H3 containing the CENP-A targeting domain. Mol. Cell 25, 309–322 (2007).

    Article  CAS  Google Scholar 

  28. Fujita, Y. et al. Priming of centromere for CENP-A recruitment by human hMis18alpha, hMis18beta, and M18BP1. Dev. Cell 12, 17–30 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Guse, A., Carroll, C. W., Moree, B., Fuller, C. J. & Straight, A. F. In vitro centromere and kinetochore assembly on defined chromatin templates. Nature 477, 354–358 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Barnhart, M. C. et al. HJURP is a CENP-A chromatin assembly factor sufficient to form a functional de novo kinetochore. J. Cell Biol. 194, 229–243 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mendiburo, M. J., Padeken, J., Fulop, S., Schepers, A. & Heun, P. Drosophila CENH3 is sufficient for centromere formation. Science 334, 686–690 (2011).

    Article  CAS  Google Scholar 

  32. 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 

  33. 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 

  34. Russell, D. W. & Hirata, R. K. Human gene targeting by viral vectors. Nat. Genet. 18, 325–330 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Berdougo, E., Terret, M. E. & Jallepalli, P. V. Functional dissection of mitotic regulators through gene targeting in human somatic cells. Methods Mol. Biol. 545, 21–37 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Black, B. E. et al. Structural determinants for generating centromeric chromatin. Nature 430, 578–582 (2004).

    Article  CAS  Google Scholar 

  37. Carroll, C. W., Milks, K. J. & Straight, A. F. Dual recognition of CENP-A nucleosomes is required for centromere assembly. J. Cell Biol. 189, 1143–1155 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Carroll, C. W., Silva, M. C., Godek, K. M., Jansen, L. E. & Straight, A. F. Centromere assembly requires the direct recognition of CENP-A nucleosomes by CENP-N. Nat. Cell Biol. 11, 896–902 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Foltz, D. R. et al. The human CENP-A centromeric nucleosome-associated complex. Nat. Cell Biol. 8, 458–469 (2006).

    Article  CAS  Google Scholar 

  40. Nishino, T. et al. CENP-T-W-S-X forms a unique centromeric chromatin structure with a histone-like fold. Cell 148, 487–501 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hori, T. et al. CCAN makes multiple contacts with centromeric DNA to provide distinct pathways to the outer kinetochore. Cell 135, 1039–1052 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Okada, M. et al. The CENP-H-I complex is required for the efficient incorporation of newly synthesized CENP-A into centromeres. Nat. Cell Biol. 8, 446–457 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Cheeseman, I. M., Chappie, J. S., Wilson-Kubalek, E. M. & Desai, A. The conserved KMN network constitutes the core microtubule-binding site of the kinetochore. Cell 127, 983–997 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Masumoto, H., Masukata, H., Muro, Y., Nozaki, N. & Okazaki, T. A human centromere antigen (CENP-B) interacts with a short specific sequence in alphoid DNA, a human centromeric satellite. J. Cell Biol. 109, 1963–1973 (1989).

    Article  CAS  PubMed  Google Scholar 

  45. 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  Google Scholar 

  46. 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 

  47. Bassett, E. A. et al. HJURP uses distinct CENP-A surfaces to recognize and to stabilize CENP-A/histone H4 for centromere assembly. Dev. Cell 22, 749–762 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hu, H. et al. Structure of a CENP-A-histone H4 heterodimer in complex with chaperone HJURP. Genes Dev. 25, 901–906 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Panchenko, T. et al. Replacement of histone H3 with CENP-A directs global nucleosome array condensation and loosening of nucleosome superhelical termini. Proc. Natl Acad. Sci. USA 108, 16588–16593 (2011).

    Article  PubMed  Google Scholar 

  50. 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 

  51. Castillo, A. G. et al. Plasticity of fission yeast CENP-A chromatin driven by relative levels of histone H3 and H4. PLoS Genet. 3, 1264–1274 (2007).

    Article  CAS  Google Scholar 

  52. Torras-Llort, M., Moreno-Moreno, O. & Azorin, F. Focus on the centre: the role of chromatin on the regulation of centromere identity and function. EMBO J. 28, 2337–2348 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ribeiro, S. A. et al. A super-resolution map of the vertebrate kinetochore. Proc. Natl Acad. Sci. USA 107, 10484–10489 (2010).

    Article  PubMed  Google Scholar 

  54. Black, B. E., Brock, M. A., Bedard, S., Woods, V. L. Jr & Cleveland, D. W. An epigenetic mark generated by the incorporation of CENP-A into centromeric nucleosomes. Proc. Natl Acad. Sci. USA 104, 5008–5013 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Trazzi, S. et al. The C-terminal domain of CENP-C displays multiple and critical functions for mammalian centromere formation. PLoS ONE 4, e5832 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sugimoto, K., Yata, H., Muro, Y. & Himeno, M. Human centromere protein C (CENP-C) is a DNA-binding protein which possesses a novel DNA-binding motif. J. Biochem. 116, 877–881 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. Suzuki, N. et al. CENP-B interacts with CENP-C domains containing Mif2 regions responsible for centromere localization. J. Biol. Chem. 279, 5934–5946 (2004).

    Article  CAS  PubMed  Google Scholar 

  58. Okada, T. et al. CENP-B controls centromere formation depending on the chromatin context. Cell 131, 1287–1300 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Sullivan, B. A. & Karpen, G. H. Centromeric chromatin exhibits a histone modification pattern that is distinct from both euchromatin and heterochromatin. Nat. Struct. Mol. Biol. 11, 1076–1083 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hasson, D. et al. The octamer is the major form of CENP-A nucleosomes at human centromeres. Nat. Struct. Mol. Biol. 20, 687–695 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tanaka, Y. et al. Human centromere protein B induces translational positioning of nucleosomes on alpha-satellite sequences. J. Biol. Chem. 280, 41609–41618 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. Okamoto, Y., Nakano, M., Ohzeki, J., Larionov, V. & Masumoto, H. A minimal CENP-A core is required for nucleation and maintenance of a functional human centromere. EMBO J. 26, 1279–1291 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sakuno, T., Tada, K. & Watanabe, Y. Kinetochore geometry defined by cohesion within the centromere. Nature 458, 852–858 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Shah, J. V. et al. Dynamics of centromere and kinetochore proteins;implications for checkpoint signaling and silencing. Curr. Biol. 14, 942–952 (2004).

    CAS  PubMed  Google Scholar 

  65. Bodor, D. L., Rodriguez, M. G., Moreno, N. & Jansen, L. E. Analysis of protein turnover by quantitative SNAP-based pulse-chase imaging. Curr. Protoc. Cell Biol. Chapter 8, Unit8 8 (2012).

  66. Moreno, S., Klar, A. & Nurse, P. Molecular genetic analysis offission yeast Schizosaccharomyces pombe. Methods Enzymol. 194, 795–823 (1991).

    Article  CAS  PubMed  Google Scholar 

  67. 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 

Download references

Acknowledgements

The authors would like to thank P. Jallepalli (Sloan-Kettering, New York, USA) for helpful suggestions, J. F. Mata and M. C. C. Silva (Gulbenkian Institute, Oeiras, Portugal), T. Panchenko (University of Pennsylvania, Philadelphia, USA) for technical help, D. Foltz (University of Virginia, Charlottesville, USA), A. Straight (Stanford University, USA), P. Maddox (Université de Montréal, Canada), S-T. Liu (University of Toledo, USE), A. Miyawaki (RIKEN, Japan), R. Allshire (WTCCB, Edinburgh, UK), Y. Watanabe (University of Tokyo, Japan) and O. Limbo and P. Russell (TSRI, La Jolla, USA) for providing reagents. We also thank B. E. Black and C. Bartocci (TSRI, La Jolla) for helpful comments on the manuscript, E. Khaliullina for drawing the model in Fig. 8, the Neuroscience Microscopy Shared Facility (P30 NS047101, University of California, San Diego) and the FACS facility in the HESCCF (Sanford Consortium for Regenerative Medicine, La Jolla, CA). This work was supported by a grant (GM 074150) from the National Institutes of Health to D.W.C., who receives salary support from the Ludwig Institute for Cancer Research. D.F. was supported by a European Molecular Biology Organization (EMBO) long-term fellowship.

Author information

Authors and Affiliations

Authors

Contributions

H.D.F. and A.D. designed and performed yeast experiments and contributed to manuscript writing. L.P.V. and L.E.T.J. performed FISH experiments. L.E.T.J. targeted the first CENP-A allele. D.F., Y.N-A., K.N., A.J.W., Q.Z., A.J.H. performed the experiments. D.F. analysed the data. D.F. and D.W.C. conceived the experimental design and wrote the manuscript.

Corresponding author

Correspondence to Don W. Cleveland.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 CENP-A conditional disruption in human cells.

(a) CENP-A genotypes validated in the indicated cell lines using PCR to distinguish normal (+), disrupted (-), and floxed (F) alleles, as diagramed to right. (b) To monitor growth rate, CENP-A+/+, CENP-AF/+ and CENP-A−/F RPE1 cells were counted every 2 days. Values represent the mean of three independent experiments, and error bars represent the SEM. (c) Immunoblot to determine CENP-A levels in CENP-A+/+, CENP-AF/+ and CENP-A−/F RPE1 cells. (d) Representative fluorescence images of centromeric CENP-A levels. Bar graphs show CENP-A protein intensity in the indicated cell lines. Bars represent the mean of three independent experiments (>30 cells per experiment). Error bars represent the SEM. ACA staining was used to identify centromeres in CENP-AF/+ and CENP-A−/F RPE1 cells. (e) Efficiency of excision of exons 3 and 4 in the overall cell population determined by Q-PCR after Ad-Cre transduction of CENP-A−/F RPE1 cells. Position of the primers is illustrated in the schematic at top. (f) PCR analysis on the DNA extracted from the survival colonies in the CENP-A−/F RPE1 cells after Ad-CRE treatment. Images show representative crystal violet-stained colonies for the indicated cell lines with or without treatment with Ad-CRE. (g) Schematic of introduction of the LoxPRFP-STOPLoxP-GFP gene by retroviral integration into CENP-A−/F cells, followed by FACS sorting to isolate only green fluorescing cells with both CENP-A alleles inactivated. (h) Graph shows the growth rate of CENP-AF/+ and CENP-A−/F with RFP-STOP-GFP construct of the experiment described in f. A parallel-untreated control is also shown. Cells were counted every 2 days. Values represent the mean of three independent experiments, and error bars represent the SEM. Scale bar = 5 μm.

Supplementary Figure 2 Centromeric proteins dissociate from centromeres with different kinetics during CENP-A depletion.

(a) Representative fluorescence images show the localization and intensity of CENP-P (purple bars). ACA staining was used to identify centromeres. Red bars show CENP-A intensity from Fig. 1e. Bars represent the mean of three independent experiments (>30 cells per experiment). Error bars represent the SEM. CENP-P was tracked by covalent labeling with rhodamine-benzyl guanine after stable expression of a gene encoding SNAP-tagged CENP-P (b) As in A, except that CENP-C and Ndc80 at day 0 and 7 are shown after Ad-Cre infection. Cells were arrested in monostarol for 12 hours. Yellow bars show Ndc80 intensity of the fluorescence images at the indicated days after Ad-Cre treatment. (c) As in a, but for CENP-A and CENP-I levels at day 0 and 9 after Ad-Cre infection. Interphase cells were quantified. Statistics source data are in Supplementary Table S2. Scale bar = 5 μm.

Supplementary Figure 3 Clonogenic and immunofluorescence localization assays determine that the CATD is sufficient for centromere identity and for templating its replication.

(a) CENP-A amino acid sequence and the domains, including the amino terminal tail (red), the αN helix (green), the CATD (light blue), and the carboxyl terminal tail (orange). (b) Representative fluorescence images of the localization of EYFP-rescue constructs of experiment outlined in Fig. 3. (c) Clongenic survival assays (as in Fig. 3d) for the CENP-A−/F cells with and without incubation in Ad-Cre and with the indicated rescue constructs. (d) Representative fluorescence images of the localization and intensity of CENP-T, CENP-A and EYFP-H3CATD at the indicated days after Ad-Cre treatment. Scale bar = 5 μm.

Supplementary Figure 4 H3CATD combined with either the amino or carboxyl tails of human CENP-A confer long term centromere identify and kinetochore function.

(a) (Top) Clonogenic survival assays for RPE1 cells with the indicated CENP-A genotypes and rescue constructs. (Bottom) Corresponding growth rates determined by counting cell numbers every 2 days for CENP-A−/F cells and CENP-A−/− cells with CENP-A, NH2H3CATD, or H3CATD−C rescue genes. Values represent the mean of three independent experiments, and error bars represent the SEM. (b) Confirmation of excision of endogenous CENP-A alleles in the indicated survival rescue construct cells. A schematic representation of the position of the primers is also shown. (c) (Top) Representative fluorescence images show the loss of endogenous CENP-A at centromeres only for cells lacking the amino-terminus of CENP-A epitopes recognized by our CENP-A antibody. ACA staining was used to identify centromeres. (Bottom) Quantitation of the fluorescence images in the CENP-A−/−+H3CATD−C and CENP-A−/−+NH2CENP-A cells. (d) Representative fluorescence images showing localization of the NH2H3CATDSNAP in CENP-A−/− cells during different phases of the cell cycle. ACA staining was used to identify centromeres and a-tubulin to identify telophase/early G1 cells. (e) Indirect immunofluorescence images of localization and intensity of EYFP-H3CATD−C at centromeres after GAPDH or HJURP siRNA, respectively. ACA staining was used to identify centromeres. (f) Quantitation of EYFP intensity at centromeres in images from (e) after GAPDH, HJURP siRNA, or no siRNA. Error bars represent the SEM of two independent experiments (>30 cells per experiment). ACA staining was used to identify centromeres. (g) (Left) CENP-C protein intensity after GAPDH or CENP-C siRNA in the indicated cell lines, respectively. Bars represent the mean of two independent experiments (>30 cells per experiment). (Right) Percentage of cells that undergo division after 72 hours of CENP-C depletion, determined by live cell microscopy. Error bars represent the SEM. Statistics source data are in Supplementary Table S2. Scale bar = 5 μm

Supplementary Figure 5 Accurate chromosome segregation with centromere-targeted histone H3 carrying either CENP-A tail domain.

(a) Representative fluorescence images of localization and intensity of centromere-bound CENP-I or Ndc80 in CENP-A−/− cell lines with the indicated rescue genes. ACA staining (red) is also shown. (b) Representative fluorescence images showing localization and intensity of centromere bound CENP-B in the indicated cell lines. (c) Representative fluorescence images showing localization and intensity of CENP-B in the indicated cell lines after 2 days of shRNA against CENP-B. (Right) Quantitation of CENP-B protein intensity of fluorescence images. (d) Time-lapse images of CENP-A−/−+ EYFPNH2H3CATD (green) RPE1 cells also stably expressing histone H2B-mRFP (red). Numbers at the top right refer to the time in minutes after nuclear envelope breakdown (NEBD). Yellow arrows show the formation of micronuclei. (e) (Left) Quantitation of misaligned or lagging chromosomes or subsequent micronuclei formation or (right) mitotic timing, determined from images in (d). Time in mitosis was defined by the period from NEBD to chromosome decondensation. Bars represent the mean of >50 cells per condition. Each individual point represents a single cell. Error bars represent the SEM. Statistics source data are in Supplementary Table S2. Scale bar = 5 μm.

Supplementary Figure 6 Protein level expression of the fission yeast rescue constructs.

Immunoblot to quantify levels of each GFP-tagged rescue variant, visualized with a GFP antibody. β-actin is used as a loading control.

Supplementary Figure 7 Uncropped image of blots from Figs 1c, 3c, 4d, 5c, 6b.

Supplementary Table 1 List of strains used in this study. Fission yeast strains used in Fig. 7 are listed. The source column indicates where the original strain comes from. The genotype of each strain is also indicated. [68Pidoux, 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)].
Supplementary Table 2 Statistic source data (n<5).

Supplementary information

Supplementary Information

Supplementary Information (PDF 1056 kb)

Supplementary Table 1

Supplementary Information (XLSX 51 kb)

Supplementary Table 2

Supplementary Information (XLSX 49 kb)

A method to eliminate Cre-escaper cells.

Movie of CENP-A−/F cells expressing the LoxPRFP-STOPLoxP-GFP cassette one hour after infection with Ad-Cre. One frame captured every 5 minutes. Time at the top left shows number of minutes after the start of filming. (MOV 12652 kb)

Lowered levels of CENP-A corresponded with increased rates of chromosome segregation defects.

Movie of CENP-A−/F cells expressing Histone H2B-mRFP five days after infection with Ad-Cre. One frame captured every 3 minutes. (AVI 2196 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fachinetti, D., Diego Folco, H., Nechemia-Arbely, Y. et al. A two-step mechanism for epigenetic specification of centromere identity and function. Nat Cell Biol 15, 1056–1066 (2013). https://doi.org/10.1038/ncb2805

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2805

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