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:

Higher-order protein assembly controls kinetochore formation

Abstract

To faithfully segregate chromosomes during vertebrate mitosis, kinetochore–microtubule interactions must be restricted to a single site on each chromosome. Prior work on pair-wise kinetochore protein interactions has been unable to identify the mechanisms that prevent outer kinetochore formation in regions with a low density of CENP-A nucleosomes. To investigate the impact of higher-order assembly on kinetochore formation, we generated oligomers of the inner kinetochore protein CENP-T using two distinct, genetically engineered systems in human cells. Although individual CENP-T molecules interact poorly with outer kinetochore proteins, oligomers that mimic centromeric CENP-T density trigger the robust formation of functional, cytoplasmic kinetochore-like particles. Both in cells and in vitro, each molecule of oligomerized CENP-T recruits substantially higher levels of outer kinetochore components than monomeric CENP-T molecules. Our work suggests that the density dependence of CENP-T restricts outer kinetochore recruitment to centromeres, where densely packed CENP-A recruits a high local concentration of inner kinetochore proteins.

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

Fig. 1: I3-01 oligomerization strategy generates particles that interact with mitotic spindles.
Fig. 2: CENP-T1–242 oligomers recruit almost the entire outer kinetochore.
Fig. 3: Isolated CENP-T1–242 oligomers bind to microtubules and track dynamic microtubule ends.
Fig. 4: CENP-T1–242 oligomerization promotes outer kinetochore recruitment.
Fig. 5: Each additional CENP-T1–242 molecule incrementally increases outer kinetochore recruitment of neighbouring molecules.
Fig. 6: Oligomerization of CENP-T is required to saturate NDC80 binding sites.

Similar content being viewed by others

Data availability

The proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE90 partner repository with the dataset identifiers PXD042174 and https://doi.org/10.6019/PXD042174. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Cheeseman, I. M. The kinetochore. Cold Spring Harb. Perspect. Biol. https://doi.org/10.1101/cshperspect.a015826 (2014).

  2. Musacchio, A. & Desai, A. A molecular view of kinetochore assembly and function. Biology 6, 5 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Suzuki, A., Badger, B. L. & Salmon, E. D. A quantitative description of Ndc80 complex linkage to human kinetochores. Nat. Commun. https://doi.org/10.1038/ncomms9161 (2015).

  4. Brinkley, B. R. & Cart Wright, J. Ultrastructural analysis of mitotic spindle elongation in mammalian cells in vitro. Direct microtubule counts. J. Cell Biol. 50, 416–431 (1971).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhou, K. et al. CENP-N promotes the compaction of centromeric chromatin. Nat. Struct. Mol. Biol. 29, 403–413 (2022).

  6. Hara, M. et al. Centromere/kinetochore is assembled through CENP-C oligomerization. Mol. Cell https://doi.org/10.1016/j.molcel.2023.05.023 (2023).

  7. Cheeseman, I. M. & Desai, A. Molecular architecture of the kinetochore–microtubule interface. Nat. Rev. Mol. Cell Biol. 9, 33–46 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Xia, S. et al. Higher-order assemblies in immune signaling: supramolecular complexes and phase separation. Protein Cell 12, 680 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wu, H. Higher-order assemblies in a new paradigm of signal transduction. Cell 153, 287–292 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wu, H. & Fuxreiter, M. The structure and dynamics of higher-order assemblies: amyloids, signalosomes, and granules. Cell 165, 1055–1066 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Banani, S. F. et al. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18, 285–298 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Marzahn, M. R. et al. Higher-order oligomerization promotes localization of SPOP to liquid nuclear speckles. EMBO J. 35, 1254–1275 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Navarro, A. P. & Cheeseman, I. M. Kinetochore assembly throughout the cell cycle. Semin. Cell Dev. Biol. 117, 62–74 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Gascoigne, K. E. & Cheeseman, I. M. CDK-dependent phosphorylation and nuclear exclusion coordinately control kinetochore assembly state. J. Cell Biol. 201, 23–32 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Rago, F., Gascoigne, K. E. & Cheeseman, I. M. Distinct organization and regulation of the outer kinetochore KMN network downstream of CENP-C and CENP-T. Curr. Biol. 25, 671–677 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hara, M. et al. Multiple phosphorylations control recruitment of the KMN network onto kinetochores. Nat. Cell Biol. 20, 1378–1388 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Kim, S. & Yu, H. Multiple assembly mechanisms anchor the KMN spindle checkpoint platform at human mitotic kinetochores. J. Cell Biol. 208, 181–196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nishino, T. et al. CENP-T provides a structural platform for outer kinetochore assembly. EMBO J. 32, 424–436 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Huis In’t Veld, P. J. et al. Molecular basis of outer kinetochore assembly on CENP-T. eLife https://doi.org/10.7554/eLife.21007 (2016).

    Article  PubMed  Google Scholar 

  20. Palladino, J. et al. Targeted de novo centromere formation in Drosophila reveals plasticity and maintenance potential of CENP-A chromatin. Dev. Cell 52, 379–394.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gascoigne, K. E. et al. Induced ectopic kinetochore assembly bypasses the requirement for CENP-A nucleosomes. Cell https://doi.org/10.1016/j.cell.2011.03.031 (2011).

  22. Gascoigne, K. E. & Cheeseman, I. M. Induced dicentric chromosome formation promotes genomic rearrangements and tumorigenesis. Chromosome Res. 21, 407–418 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mckinley, K. L. & Cheeseman, I. M. The molecular basis for centromere identity and function. Nat. Publ. Group 17, 16–29 (2015).

    Google Scholar 

  24. 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).

    Article  PubMed  Google Scholar 

  25. Walstein, K. et al. Assembly principles and stoichiometry of a complete human kinetochore module. Sci. Adv. 7, 27 (2021).

  26. Yatskevich, S. et al. Structure of the human inner kinetochore bound to a centromeric CENP-A nucleosome. Science 376, 844–852 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pesenti, M. E. et al. Structure of the human inner kinetochore CCAN complex and its significance for human centromere organization. Mol. Cell 82, 2113–2131.e8 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tian, T. et al. Structural insights into human CCAN complex assembled onto DNA. Cell Discov. 8, 1–15 (2022).

    Article  CAS  Google Scholar 

  29. Tarasovetc, E. V. et al. Permitted and restricted steps of human kinetochore assembly in mitotic cell extracts. Mol. Biol. Cell 32, 1241–1255 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Screpanti, E. et al. Direct binding of Cenp-C to the Mis12 complex joins the inner and outer kinetochore. Curr. Biol. 21, 391–398 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Petrovic, A. et al. The MIS12 complex is a protein interaction hub for outer kinetochore assembly. J. Cell Biol. 190, 835–852 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Drinnenberg, I. A., Henikoff, S. & Malik, H. S. Evolutionary turnover of kinetochore proteins: a ship of Theseus? Trends Cell Biol. 26, 498–510 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Takenoshita, Y., Hara, M. & Fukagawa, T. Recruitment of two Ndc80 complexes via the CENP-T pathway is sufficient for kinetochore functions. Nat. Commun. 13, 1–19 (2022).

    Article  Google Scholar 

  34. 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 

  35. Hsia, Y. et al. Design of a hyperstable 60-subunit protein icosahedron. Nature 535, 136–139 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. 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 

  37. Musacchio, A. The molecular biology of spindle assembly checkpoint signaling dynamics. Curr. Biol. 25, R1002–R1018 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Monda, J. K. & Cheeseman, I. M. The kinetochore–microtubule interface at a glance. J. Cell Sci. 131, jcs214577 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kops, G. J. P. L. & Gassmann, R. Crowning the kinetochore: the fibrous corona in chromosome segregation. Trends Cell Biol. 30, 653–667 (2020).

    Article  CAS  PubMed  Google Scholar 

  40. Grishchuk, E. L. Biophysics of microtubule end coupling at the kinetochore. Prog. Mol. Subcell. Biol. 56, 397–428 (2017).

    Article  CAS  PubMed  Google Scholar 

  41. Maiato, H. et al. Mechanisms of chromosome congression during mitosis. Biology 6, 13 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Cheeseman, I. M. et al. The conserved KMN network constitutes the core microtubule-binding site of the kinetochore. Cell 127, 983–997 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Cai, S. et al. Chromosome congression in the absence of kinetochore fibres. Nat. Cell Biol. 11, 832–838 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hunt, A. J. & McIntosh, J. R. The dynamic behavior of individual microtubules associated with chromosomes in vitro. Mol. Biol. Cell 9, 2857–2871 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Volkov, V. A. et al. Multivalency of NDC80 in the outer kinetochore is essential to track shortening microtubules and generate forces. eLife 7, e36764 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zaytsev, A. V., Ataullakhanov, F. I. & Grishchuk, E. L. Highly transient molecular interactions underlie the stability of kinetochore–microtubule attachment during cell division. Cell. Mol. Bioeng. 6, 393–405 (2013).

    Article  CAS  Google Scholar 

  47. Tanenbaum, M. E. et al. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ciferri, C. et al. Implications for kinetochore–microtubule attachment from the structure of an engineered Ndc80 complex. Cell 133, 427–439 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Itzhak, D. N. et al. Global, quantitative and dynamic mapping of protein subcellular localization. eLife 5, e16950 (2016).

  50. Wiśniewski, J. R. et al. A “proteomic ruler” for protein copy number and concentration estimation without spike-in standards. Mol. Cell. Proteom. 13, 3497–3506 (2014).

    Article  Google Scholar 

  51. Schmidt, J. C. et al. The kinetochore-bound Ska1 complex tracks depolymerizing microtubules and binds to curved protofilaments. Dev. Cell 23, 968–980 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zaytsev, A. V. et al. Accurate phosphoregulation of kinetochore–microtubule affinity requires unconstrained molecular interactions. J. Cell Biol. 206, 45–59 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. McKinley, K. L. & Cheeseman, I. M. The molecular basis for centromere identity and function. Nat. Rev. Mol. Cell Biol. 17, 16–29 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Nechemia-Arbely, Y. et al. DNA replication acts as an error correction mechanism to maintain centromere identity by restricting CENP-A to centromeres. Nat. Cell Biol. 21, 743 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Athwal, R. K. et al. CENP-A nucleosomes localize to transcription factor hotspots and subtelomeric sites in human cancer cells. Epigenet. Chromatin 8, 23 (2015).

    Article  Google Scholar 

  56. 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 

  57. De Rop, V., Padeganeh, A. & Maddox, P. S. CENP-A: the key player behind centromere identity, propagation, and kinetochore assembly. Chromosoma 121, 527 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bodor, D. L. et al. The quantitative architecture of centromeric chromatin. eLife 3, e02137 (2014).

  59. Akiyoshi, B. et al. Quantitative proteomic analysis of purified yeast kinetochores identifies a PP1 regulatory subunit. Genes Dev. 23, 2887–2899 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Akiyoshi, B. et al. Tension directly stabilizes reconstituted kinetochore–microtubule attachments. Nature 468, 576–579 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Miller, M. P., Asbury, C. L. & Biggins, S. A TOG protein confers tension sensitivity to kinetochore–microtubule attachments. Cell 165, 1428–1439 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gonen, S. et al. The structure of purified kinetochores reveals multiple microtubule-attachment sites. Nat. Struct. Mol. Biol. 19, 925–929 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Powers, A. F. et al. The Ndc80 kinetochore complex forms load-bearing attachments to dynamic microtubule tips via biased diffusion. Cell 136, 865–875 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Helgeson, L. A. et al. Human Ska complex and Ndc80 complex interact to form a load-bearing assembly that strengthens kinetochore–microtubule attachments. Proc. Natl Acad. Sci. USA 115, 2740–2745 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Huis in’T Veld, P. J. et al. Molecular determinants of the Ska–Ndc80 interaction and their influence on microtubule tracking and force-coupling. eLife 8, e49539 (2019).

  66. Luo, W. et al. CLASP2 recognizes tubulins exposed at the microtubule plus-end in a nucleotide state-sensitive manner. Sci. Adv. 9, eabq5404 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Chakraborty, M. et al. Microtubule end conversion mediated by motors and diffusing proteins with no intrinsic microtubule end-binding activity. Nat. Commun. 10, 1–14 (2019).

    Article  Google Scholar 

  68. Hori, T. et al. 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 

  69. Bhat, P., Honson, D. & Guttman, M. Nuclear compartmentalization as a mechanism of quantitative control of gene expression. Nat. Rev. Mol. Cell Biol. 22, 653–670 (2021).

    Article  CAS  PubMed  Google Scholar 

  70. Li, P. et al. Phase transitions in the assembly of multivalent signalling proteins. Nature 483, 336–340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Schwarz-Romond, T. et al. The DIX domain of Dishevelled confers Wnt signaling by dynamic polymerization. Nat. Struct. Mol. Biol. 14, 484–492 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Li, J. et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Lu, A. et al. Plasticity in PYD assembly revealed by cryo-EM structure of the PYD filament of AIM2. Cell Discov. 1, 1–14 (2015).

    Article  Google Scholar 

  74. Lu, A. & Wu, H. Structural mechanisms of inflammasome assembly. FEBS J. 282, 435–444 (2015).

    Article  CAS  PubMed  Google Scholar 

  75. Park, H. et al. Optogenetic protein clustering through fluorescent protein tagging and extension of CRY2. Nat. Commun. 8, 1–8 (2017).

    Article  Google Scholar 

  76. Bugaj, L. J. et al. Optogenetic protein clustering and signaling activation in mammalian cells. Nat. Methods 10, 249–252 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Qian, K. et al. A simple and efficient system for regulating gene expression in human pluripotent stem cells and derivatives. Stem Cells 32, 1230–1238 (2014).

    Article  CAS  PubMed  Google Scholar 

  78. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science 350, 1096–1101 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. McKinley, K. L. & Cheeseman, I. M. Large-scale analysis of CRISPR/Cas9 cell-cycle knockouts reveals the diversity of p53-dependent responses to cell-cycle defects. Dev. Cell https://doi.org/10.1016/j.devcel.2017.01.012 (2017).

  81. Welburn, J. P. I. et al. The human kinetochore Ska1 complex facilitates microtubule depolymerization-coupled motility. Dev. Cell 16, 374–385 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Cheeseman, I. M. & Desai, A. A combined approach for the localization and tandem affinity purification of protein complexes from metazoans. Sci. STKE 2005, 266 (2005).

  83. Cheeseman, I. M. et al. KNL1 and the CENP-H/I/K complex coordinately direct kinetochore assembly in vertebrates. Mol. Biol. Cell 19, 587–594 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  85. Stirling, D. R. et al. CellProfiler 4: improvements in speed, utility and usability. BMC Bioinform. 22, 1–11 (2021).

    Article  Google Scholar 

  86. Volkov, V. A., Zaytsev, A. V. & Grishchuk, E. L. Preparation of segmented microtubules to study motions driven by the disassembling microtubule ends. J. Vis. Exp. 85, 51150 (2014).

    Google Scholar 

  87. Miller, H. P. & Wilson, L. Preparation of microtubule protein and purified tubulin from bovine brain by cycles of assembly and disassembly and phosphocellulose chromatography. Methods Cell Biol. 95, 2–15 (2010).

    Article  Google Scholar 

  88. Hyman, A. A. & Mitchison, T. J. Two different microtubule-based motor activities with opposite polarities in kinetochores. Nature 351, 206–211 (1991).

    Article  CAS  PubMed  Google Scholar 

  89. Chakraborty, M., Tarasovetc, E. V. & Grishchuk, E. L. in Mitosis and Meiosis Part A (eds Maiato, H. & Schuh, M.) Ch. 13 (2018).

  90. Perez-Riverol, Y. et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 50, D543–D552 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Ly and S. Bell for feedback on the paper, and members of the Cheeseman and Grishchuk labs for feedback throughout the process. This work was supported by grants to I.M.C. from the NIH/NIGMS (R35GM126930), including diversity supplement funding for G.B.S., and the Gordon and Betty Moore Foundation, a grant to E.L.G. from NIH/NIGMS (R35-GM141747), and grants to both I.M.C. and E.L.G. from the American Cancer Society Theory Lab Collaborative Grant (TLC-20-117-01-TLC) and NSF (2029868).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization—G.B.S., I.M.C. and E.L.G.; methodology, validation and investigation—G.B.S. for reagent generation, experimental design and all in vivo experiments, E.V.T. for all in vitro experiments, and O.M. for western blots and additional support; writing, original draft preparation—G.B.S. and I.M.C.; writing, review and editing—E.L.G., E.V.T. and O.M.; visualization—G.B.S., E.V.T. and O.M.; supervision—I.M.C. and E.L.G.; funding acquisition—I.M.C. and E.L.G.

Corresponding authors

Correspondence to Ekaterina L. Grishchuk or Iain M. Cheeseman.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Kerry Bloom and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 CENP-T1–242 oligomers interact with spindles and recruit additional outer kinetochore proteins, but control oligomers do not.

(a) Co-localization of outer kinetochore proteins with GFP-CENP-T1–242-I3-01 oligomers by immunofluorescence. Identical linear brightness adjustments were used for GFP and kinetochore protein channels for each pair of experimental and control samples. Regions enlarged in insets are indicated by dashed boxes. Full-size image scale bars=5 µm. Inset scale bars=2 µm. SKA3 experiment was repeated 4 times with similar results. CENP-A and ZW10 experiments were repeated twice with similar results. (b) Pearson correlations between GFP and kinetochore protein signals for GFP-I3-01 and GFP-CENP-T1–242-I3-01. Each point is a cell; n=number of cells measured in a single experiment. Bars represent mean ± SEM.; each experiment was performed 2 times with similar results. Statistical analysis of replicates and sample sizes can be found in Supplementary Table 4. P-values were calculated with Welch’s two-tailed t-tests: ZW10: p < 0.0001; SKA3: p < 0.0001; CENPA: p = 0.0809. Source numerical data are available in Source Data.

Source data

Extended Data Fig. 2 CENP-T1–242 oligomers recruit additional outer kinetochore proteins, but control oligomers do not.

(a) Outer kinetochore and kinetochore-associated proteins detected in immuno-precipitation mass spectrometry of GFP-I3-01 control oligomers. This experiment was performed twice with similar results. (b) Peptides counts for inner kinetochore proteins detected in immunoprecipitation mass spectrometry of GFP-I3-01 and GFP-CENP-T1–242-I3-01 oligomers. This experiment was performed twice with similar results.

Extended Data Fig. 3 Characterization of GFP CENP-T1–242 and control GFP oligomers isolated from HeLa cells.

(a) Workflow to isolate GFP-CENP-T1–242-I3-01 and GFP-I3-01 from mitotic cells. Left: representative images of HeLa cells expressing GFP-CENP-T1–242 or GFP oligomers. Cells were arrested in mitosis by expression of GFP-CENP-T1–242-I3-01 or with the Eg5 inhibitor S-Trityl-L-Cysteine (see Methods for details). (b) Quantification of the number of GFP molecules. Left: representative image of a microscope field with single GFP molecules immobilized on plasma-cleaned coverslip. Repeated 3 times with similar results. Middle: Example photobleaching curve for a single molecule of GFP. Right: Histogram of integral intensities collected from 60 bleaching GFP dots from N = 3 independent experiments. Each point represents the frequency in one independent repeat. Red line is fit to Gaussian function. Bars represent mean ± SEM. Peak value of 1.56 ± 0.04×104 a.u. is the integral intensity of a single GFP fluorophore under our imaging conditions. This intensity was used to estimate number of GFP fluorophores in oligomers and complexes, see Methods for details. (c) Representative fluorescence microscopy images of the indicated GFP-labelled oligomers immobilized on coverslips; identical microscopy settings and brightness adjustments were used. Repeated 5 times with similar results. (d) Immunofluorescence measurements of NDC80 intensity associated with CENP-T1–242 oligomers and GFP oligomers that bound to taxol-stabilized microtubules or did not bind to microtubules. Each point represents the median value from an independent experiment. Bars represent mean ± SEM. For microtubule-bound GFP-CENP-T1–242-I3-01, N = 2; for other conditions N = 5. Two-tailed Welch’s t-test: GFP-CENP-T1–242-I3-01 unbound vs. GFP-I3-01 unbound: p = 0.2129. (e) Kymographs illustrating complex motions of CENP-T1–242 oligomers on dynamic microtubules. Top left: an CENP-T1–242 oligomer diffuses on the microtubule wall and tracks the polymerizing plus-end. Bottom left: CENP-T1–242 oligomer tracks a depolymerize end, then tracks the end when it reverts to polymerization. Right: Processive plus-end-directed movement. Velocity on the GMPCPP-containing seed (red): 0.7 μm/min; on GDP-containing lattice (blue) 3 μm/min. Plus-end directed motion was observed in 8 out of 80 total observations. Observations were made over 8 independent experiments. Source numerical data are available in Source Data.

Source data

Extended Data Fig. 4 CENP-T1–242 oligomers and monomers have distinct localization, are expressed at comparable levels, and do not reduce outer kinetochore protein expression.

(a) Pearson correlation and Manders overlap coefficient for GFP and α-tubulin co-localization in cells expressing GFP-CENP-T1–242 or GFP-CENP-T1–242-I3-01. Datapoints are cells from a single experiment. Bars represent mean ± SEM. Each experiment was performed 2 times with similar results. Statistical analysis of replicates can be found in Supplementary Table 4. Two-tailed Welch’s t-tests: Pearson correlation: p = 0.0793; Overlap: p < 0.0001. (b) Normalized GFP signals from GFP-positive cells analyzed for DNA content in Fig. 4e as measured by flow cytometry. Each point is the mean GFP signal from 3 independent experiments. Bars represent mean ± SEM. The same cell lines were used for other assays with these three constructs. (c) Histograms showing the distribution of GFP expression levels in cells from cell line in (b) as measured by flow cytometry. Repeated 3 times with similar results. (d) Western Blot for expression levels of the NDC80 complex component NDC80 in cells expressing different constructs. NDC80 was detected using an antibody against the whole complex. Anti-GFP antibody was used to show expression of the expected construct in each cell line. Beta-Actin was used as a loading control. The NDC80 complex is an obligate complex, so depletion of one component, Spc24, with siRNA resulted in a reduction in NDC80 levels. This experiment was repeated 3 times with similar results. Source numerical data and unprocessed blots are available in Source Data.

Source data

Extended Data Fig. 5 CENP-T1–242 oligomers recruit kinetochore-associated proteins and spindle assembly checkpoint proteins more robustly than monomeric CENP-T1–242.

(a) Comparison of protein co-immunoprecipitation by CENP-T1–242 oligomers and control GFP oligomers by quantitative mass spectrometry. Each point represents a biological replicate from a single multiplexed mass spectrometry run. Bars represent mean ± SEM. Analysis details can be found in Methods. (b) Comparison of protein co-immuno-precipitation by CENP-T1–242 oligomers and CENP-T1–242 monomers by TMT-based quantitative immune-precipitation mass spectrometry. Presented and analyzed as described in (a). Two-tailed Welch’s t-test: Astrin-SKAP: p = 0.2383; Spindly: p = 0.0094; Mad2L1: p = 0.0002; Bub1: p = 0.0506; Bub3: p = 0.1151; chTOG: p = 0.001. (c) Comparison of protein co-immuno-precipitation by control GFP oligomers and CENP-T1–242 monomers by TMT-based quantitative immuno-precipitation mass spectrometry. Presented and analyzed as described in (a). Source numerical data are available in Source Data.

Source data

Extended Data Fig. 6 Additional SunTag mass spectrometry, centromere depletion, and controls.

(a) Normalized GFP signals from SunTag cells in Fig. 5c as measured by flow cytometry. Each point is the mean from N = 3 independent experiments. Bars represent mean ± SEM. (b) Anti-GFP western blot of cell lines expressing scFv-sfGFP-CENP-T1–242 with different tdTomato-tagged GCN4pep scaffolds. β-Actin was used as a loading control here and in all subsequent western blots. (c) Same analysis as in (a) for tdTomato. (d) Anti-T2A western blots of SunTag cell lines with different tdTomato-tagged GCN4pep scaffolds. Anti-T2A antibody binds to the C-terminus of the scaffolds. Experiments in panels (a-d) were performed on cells from Fig. 5c. (e) and (f) Anti-RFP and Anti-GFP westerns blots of SunTag cell lines expressing tdTomato-tagged GCN4pep scaffolds used in Fig. 5d, Extended Data Fig. 6h. (g) Validation western of anti-mCherry antibodies for immunoprecitation. IN=Input, IP=Immunoprecipitation, FT=Flow-through. (h) Comparison of MIS12 and KNL1 complex abundances in anti-mCherry quantitative immunoprecipitation mass spectrometry with different SunTag scaffolds. Each point represents a biological replicate from 2 multiplexed experiments. Each bar represents the mean ± SEM Two-tailed Welch’s t-test: MIS12: 1 vs. 6: p = 0.1083; 6 vs. 10: p = 0.7135; 10 vs. 18: p = 0.0011; 1 vs. 18: p = 0.0008. KNL1: 1 vs. 6: p = 0.3592; 6 vs. 10: p = 0.1559; 10 vs. 18: p = 0.0605; 1 vs. 18: p = 0.003. (i) Quantification of MIS12 levels at centromeres in cells expressing the scFv-sfGFP-CENP-T1–242 with different GCN4pep scaffolds. Each point is a cell. Each bar represents the mean ± SEM. Measurements were pooled from 3 independent experiments. 1: n = 49; 2: n = 45; 3: n = 47; 4: n = 25; 6: n = 47; 8: n = 47; 10: n = 51; 12: n = 47. Two-tailed Welch’s t test: 1 v. 12: p < 0.0001. Welch’s ANOVA test: P < 0.0001. (j) and (k) Anti-NDC80 Complex and anti-GFP western blots of SunTag cell lines expressing scFv-sfGFP-CENP-T1–242 alongside different GCN4pep scaffolds. (l) Same as in (d). Experiments in panels (J-L) were performed on cells lines used in Fig. 5e, f, Extended Data Fig. 6i. Scaffolds in these cell lines lack the tdTomato tag. Source numerical data and unprocessed blots are available in Source Data.

Source data

Extended Data Fig. 7 Flowcytometry gating strategy for DNA content analysis of CENP-T1–242 SunTag oligomers.

Gating strategy to select the population of cells to be analyzed for DNA content analysis in Fig. 5c. A similar gating strategy was used in Fig. 4e without the tdTomato-Area parameter.

Extended Data Fig. 8 Known N-terminal NDC80 phosphorylation sites are required for NDC80 recruitment to oligomers.

(a) Representative images of colocalization of GFP and the NDC80 complex in cells expressing scFv-sfGFP-CENP-T1–242 with either wild-type (WT) CENP-T1–242 or CENP-T1–242 with T11A and T85A mutations (2 A). These constructs were expressed alongside 12xGCN4pep scaffolds. (b) Pearson correlations between GFP and NDC80 signal for experiment in (a). Datapoints are cells from a single experiment. Bars represent mean ± SEM. Repeated 2 times with similar results. Statistical analysis of replicates and sample sizes can be found in Source Data. Two-tailed Welch’s t-test: p < 0.0001. (c) Quantification of NDC80 levels at centromeres in cells expressing scFv-sfGFP-CENP-T1-242/2A with different GCN4pep scaffolds. Each bar represents the mean ± SEM of NDC80 signal from cells expressing the designated construct. Measurements were pooled from 2 different experiments. n=number of cells pooled from 2 independent experiments. 1: n = 27; 2: n = 33; 3: n = 28; 4: n = 27; 6: n = 31; 8: n = 30; 10: n = 31; 12: n = 33. Welch’s ANOVA: p < 0.0001. Two-tailed Welch’s t-test: 1 v. 12: p = 0.0237. (d) Anti-GFP and Anti-T2A western blots of cell lines expressing different GCN4pep scaffolds alongside scFv-sfGFP-CENP-T1-242/2A. Anti-T2A antibody binds to the C-terminus of the scaffolds. β-Actin was used as a loading control. These cell lines were used in for all experiments in the figure. This was a cell line validation experiment that was only performed once. Source numerical data and unprocessed blots are available in Source Data.

Source data

Extended Data Fig. 9 Additional fluorescence intensity quantifications for in vitro CENP-T-NDC80 binding assay using recombinant oligomers and NDC80 proteins.

(a) Top: Representative images of purified recombinant GFP-CENP-T1-242/3D-I3-01 oligomers attached to a coverslip. Bottom: histogram of the distribution of the number of GFP molecules per oligomer as a percentage of the total number of examined oligomers. Each point represents an independent measurement. Each bar represents the mean ± SEM from 3 independent experiments. Distribution mean ± SEM: 66 ± 10 GFP molecules. (b) Same as (a) for GFP-I3-01. 3 independent experiments. Distribution mean ± SEM: 44 ± 4 GFP molecules. (c) Same as (a) for GFP-CENP-T1-242/3D. 3 independent experiments. Distribution mean ± SEM: 1.23 ± 0.05 molecules. (d) Single molecule binding experiment with NDC80ΔSpc24/25. Top: Experimental workflow. Bottom: representative images of GFP-CENP-T1-242/3TD-I3-01 oligomers at each experimental stage. (e) Efficiency of NDC80Bonsai or NDC80ΔSpc24/25 recruitment to GFP-CENP-T1-242/3D-I3-01 oligomers. Bars represent mean ± SEM. Each point is the median result from 3 independent experiments with >12 oligomers. Data for GFP-CENP-T1-242/3D-I3-01 oligomer is duplicated from Fig. 6d. Two-tailed Welch’s t-test: p = 0.0054. (f) Graph of the stoichiometry of binding. Final GFP signal intensity as function of initial GFP signal intensity for individual oligomers. Each point represents the measurement for one oligomer pooled from N = 3 independent experiments per data set. GFP-CENP-T1-242/3D-I3-01+NDC80Bonsai: n = 85 Oligomers; GFP-CENP-T1-242/3D-I3-01 + NDC80ΔSpc24/25: n = 79 Oligomers; GFP-I3-01+NDC80Bonsai: n = 91 Oligomers. Data are fitted to linear functions. The slopes (± standard fitting error) correspond to the number of NDC80 molecules recruited per GFP-containing monomer for each combination of oligomer and NDC80 complex. (g) Photobleaching curve taken with identical microscope settings to those used for experiments with GFP-CENP-T1-242/3D (Fig. 6b, e, f). The number of GFP puncta per imaging field at each time point was normalized to the number at t = 0. Data were fitted to an exponential decay function to estimate the probability of bleaching during imaging time. Each point represents the mean ± SEM from N = 3 independent measurements. Dashed line indicates experimental exposure time in Fig. 6e, f. Source numerical data are available in source data.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1. Cell lines. Supplementary Table 2. Primary antibodies. Supplementary Table 3. Immunofluorescence fixation conditions. Supplementary Table 4. Primers and small interfering RNAs.

Supplementary Video 1

Kinetochore-like particle tracking a polymerizing microtubule end. This video shows a dynamic microtubule (blue) growing from the coverslip-bound GMPCPP microtubule seed (red) in the presence of soluble tubulin and GFP–CENP-T1–242–I3-01 oligomers (green) isolated from mitotic HeLa cells. The isolated oligomer with associated proteins (that is, kinetochore-like particle) binds to the end of a polymerizing microtubule and moves processively with the end as it polymerizes. This video plays at 7 fps, and events are shown at 3.5 times actual speed.

Supplementary Video 2

A kinetochore-like particle exhibits plus-end-directed processive movement. A larger green particle remains stationary, while a smaller kinetochore-like particle moves processively and unidirectionally towards the growing microtubule plus-end. Initial motion on the GMPCPP-containing microtubule seed is slow, but the particle speeds up on the GDP-containing microtubule wall (for more details, see Extended Data Fig. 3d). These plus-end-directed motions were rarer than the other types of motion. This video plays at 7 fps, and events are shown at 3.5 times actual speed.

Supplementary Video 3

Kinetochore-like particle diffusing along a microtubule wall and tracking a depolymerizing microtubule end. Video shows a dynamic microtubule interacting with two kinetochore-like particles. A larger particle associates with the microtubule wall and remains stationary for the duration of the video. A smaller particle lands on the microtubule wall at the site indicated with an arrow. The particle diffuses along the microtubule wall, then tracks the depolymerizing end of the microtubule. This video plays at 7 fps, and events are shown at 3.5 times actual speed.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sissoko, G.B., Tarasovetc, E.V., Marescal, O. et al. Higher-order protein assembly controls kinetochore formation. Nat Cell Biol 26, 45–56 (2024). https://doi.org/10.1038/s41556-023-01313-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01313-7

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