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.

  • Letter
  • Published:

Insights from biochemical reconstitution into the architecture of human kinetochores

Abstract

Chromosomes are carriers of genetic material and their accurate transfer from a mother cell to its two daughters during cell division is of paramount importance for life. Kinetochores are crucial for this process, as they connect chromosomes with microtubules in the mitotic spindle1. Kinetochores are multi-subunit complexes that assemble on specialized chromatin domains, the centromeres, that are able to enrich nucleosomes containing the histone H3 variant centromeric protein A (CENP-A)2. A group of several additional CENPs, collectively known as constitutive centromere associated network (CCAN)3,4,5,6, establish the inner kinetochore, whereas a ten-subunit assembly known as the KMN network creates a microtubule-binding site in the outer kinetochore7,8. Interactions between CENP-A and two CCAN subunits, CENP-C and CENP-N, have been previously described9,10,11, but a comprehensive understanding of CCAN organization and of how it contributes to the selective recognition of CENP-A has been missing. Here we use biochemical reconstitution to unveil fundamental principles of kinetochore organization and function. We show that cooperative interactions of a seven-subunit CCAN subcomplex, the CHIKMLN complex, determine binding selectivity for CENP-A over H3-nucleosomes. The CENP-A:CHIKMLN complex binds directly to the KMN network, resulting in a 21-subunit complex that forms a minimal high-affinity linkage between CENP-A nucleosomes and microtubules in vitro. This structural module is related to fungal point kinetochores, which bind a single microtubule. Its convolution with multiple CENP-A proteins may give rise to the regional kinetochores of higher eukaryotes, which bind multiple microtubules. Biochemical reconstitution paves the way for mechanistic and quantitative analyses of kinetochores.

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: Reconstitution of the CHIKMLN complex.
Figure 2: Selective cooperative binding of CHIKMLN to CENP-A mononucleosomes.
Figure 3: KMN and CHIKMLN connect CENP-A to microtubules.
Figure 4: Centromere and kinetochore assembly and propagation.

Similar content being viewed by others

References

  1. Foley, E. A. & Kapoor, T. M. Microtubule attachment and spindle assembly checkpoint signalling at the kinetochore. Nature Rev. Mol. Cell Biol. 14, 25–37 (2013)

    Article  CAS  Google Scholar 

  2. Fukagawa, T. & Earnshaw, W. C. The centromere: chromatin foundation for the kinetochore machinery. Dev. Cell 30, 496–508 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Izuta, H. et al. Comprehensive analysis of the ICEN (interphase centromere complex) components enriched in the CENP-A chromatin of human cells. Genes Cells 11, 673–684 (2006)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. 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 

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

    Article  CAS  PubMed  Google Scholar 

  7. DeLuca, J. G. et al. Kinetochore microtubule dynamics and attachment stability are regulated by Hec1. Cell 127, 969–982 (2006)

    Article  CAS  PubMed  Google Scholar 

  8. 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 

  9. 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 

  10. 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. Nature Cell Biol. 11, 896–902 (2009)

    Article  CAS  PubMed  Google Scholar 

  11. Kato, H. et al. A conserved mechanism for centromeric nucleosome recognition by centromere protein CENP-C. Science 340, 1110–1113 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Westermann, S. & Schleiffer, A. Family matters: structural and functional conservation of centromere-associated proteins from yeast to humans. Trends Cell Biol. 23, 260–269 (2013)

    Article  CAS  PubMed  Google Scholar 

  13. London, N. & Biggins, S. Signalling dynamics in the spindle checkpoint response. Nature Rev. Mol. Cell Biol. 15, 736–748 (2014)

    Article  CAS  Google Scholar 

  14. Catania, S. & Allshire, R. C. Anarchic centromeres: deciphering order from apparent chaos. Curr. Opin. Cell Biol. 26, 41–50 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hinshaw, S. M. & Harrison, S. C. An Iml3-Chl4 heterodimer links the core centromere to factors required for accurate chromosome segregation. Cell Reports 5, 29–36 (2013)

    Article  CAS  PubMed  Google Scholar 

  16. Lowary, P. T. & Widom, J. New DNA sequence rules for high affinity binding to histone octamer and sequence-directed nucleosome positioning. J. Mol. Biol. 276, 19–42 (1998)

    Article  CAS  PubMed  Google Scholar 

  17. Nagpal, H. et al. Dynamic changes in CCAN organization through CENP-C during cell-cycle progression. Mol. Biol. Cell 26, 3768–3776 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. McKinley, K. L. et al. The CENP-L-N complex forms a critical node in an integrated meshwork of interactions at the centromere-kinetochore interface. Mol. Cell 60, 886–898 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Basilico, F. et al. The pseudo GTPase CENP-M drives human kinetochore assembly. eLife 3, e02978 (2014)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Klare, K. et al. CENP-C is a blueprint for constitutive centromere-associated network assembly within human kinetochores. J. Cell Biol. 210, 11–22 (2015)

    Article  CAS  PubMed  Google Scholar 

  21. Herzog, F. et al. Structural probing of a protein phosphatase 2A network by chemical cross-linking and mass spectrometry. Science 337, 1348–1352 (2012)

    Article  ADS  CAS  PubMed  Google Scholar 

  22. 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 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Schleiffer, A. et al. CENP-T proteins are conserved centromere receptors of the Ndc80 complex. Nature Cell Biol. 14, 604–613 (2012)

    Article  CAS  PubMed  Google Scholar 

  25. Suzuki, A., Badger, B. L. & Salmon, E. D. A quantitative description of Ndc80 complex linkage to human kinetochores. Nature Commun. 6, 8161 (2015)

    Article  ADS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Westhorpe, F. G. & Straight, A. F. The split personality of CENP-A nucleosomes. Cell 150, 245–247 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bieniossek, C., Imasaki, T., Takagi, Y. & Berger, I. MultiBac: expanding the research toolbox for multiprotein complexes. Trends Biochem. Sci. 37, 49–57 (2012)

    Article  CAS  PubMed  Google Scholar 

  32. Hashimoto, Y., Zhang, S., Zhang, S., Chen, Y. R. & Blissard, G. W. Correction: BTI-Tnao38, a new cell line derived from Trichoplusia ni, is permissive for AcMNPV infection and produces high levels of recombinant proteins. BMC Biotechnol. 12, 12 (2012)

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dyer, P. N. et al. Reconstitution of nucleosome core particles from recombinant histones and DNA. Methods Enzymol. 375, 23–44 (2004)

    Article  CAS  PubMed  Google Scholar 

  34. Guse, A., Fuller, C. J. & Straight, A. F. A cell-free system for functional centromere and kinetochore assembly. Nature Protocols 7, 1847–1869 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shim, Y., Duan, M. R., Chen, X., Smerdon, M. J. & Min, J. H. Polycistronic coexpression and nondenaturing purification of histone octamers. Anal. Biochem. 427, 190–192 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nature Methods 6, 343–345 (2009)

    Article  CAS  PubMed  Google Scholar 

  37. Grimm, M., Zimniak, T., Kahraman, A. & Herzog, F. xVis: a web server for the schematic visualization and interpretation of crosslink-derived spatial restraints. Nucleic Acids Res. 43 (W1), W362–W369 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brown, P. H. & Schuck, P. Macromolecular size-and-shape distributions by sedimentation velocity analytical ultracentrifugation. Biophys. J. 90, 4651–4661 (2006)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Przewloka, M. R. et al. CENP-C is a structural platform for kinetochore assembly. Curr. Biol. 21, 399–405 (2011)

    Article  CAS  PubMed  Google Scholar 

  41. 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 

  42. Cohen, R. L. et al. Structural and functional dissection of Mif2p, a conserved DNA-binding kinetochore protein. Mol. Biol. Cell 19, 4480–4491 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Dambacher, S. et al. CENP-C facilitates the recruitment of M18BP1 to centromeric chromatin. Nucleus 3, 101–110 (2012)

    Article  PubMed  PubMed Central  Google Scholar 

  44. Milks, K. J., Moree, B. & Straight, A. F. Dissection of CENP-C-directed centromere and kinetochore assembly. Mol. Biol. Cell 20, 4246–4255 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wei, R. R., Al-Bassam, J. & Harrison, S. C. The Ndc80/HEC1 complex is a contact point for kinetochore-microtubule attachment. Nature Struct. Mol. Biol. 14, 54–59 (2007)

    Article  CAS  Google Scholar 

  46. Kiyomitsu, T., Iwasaki, O., Obuse, C. & Yanagida, M. Inner centromere formation requires hMis14, a trident kinetochore protein that specifically recruits HP1 to human chromosomes. J. Cell Biol. 188, 791–807 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Petrovic, A. et al. Modular assembly of RWD domains on the Mis12 complex underlies outer kinetochore organization. Mol. Cell 53, 591–605 (2014)

    Article  CAS  PubMed  Google Scholar 

  48. Hornung, P. et al. Molecular architecture and connectivity of the budding yeast Mtw1 kinetochore complex. J. Mol. Biol. 405, 548–559 (2011)

    Article  CAS  PubMed  Google Scholar 

  49. 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 

  50. Maskell, D. P., Hu, X. W. & Singleton, M. R. Molecular architecture and assembly of the yeast kinetochore MIND complex. J. Cell Biol. 190, 823–834 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ghongane, P., Kapanidou, M., Asghar, A., Elowe, S. & Bolanos-Garcia, V. M. The dynamic protein Knl1 - a kinetochore rendezvous. J. Cell Sci. 127, 3415–3423 (2014)

    CAS  PubMed  Google Scholar 

  52. Moree, B., Meyer, C. B., Fuller, C. J. & Straight, A. F. CENP-C recruits M18BP1 to centromeres to promote CENP-A chromatin assembly. J. Cell Biol. 194, 855–871 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. 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 

  55. Dunleavy, E. M., Almouzni, G. & Karpen, G. H. H3.3 is deposited at centromeres in S phase as a placeholder for newly assembled CENP-A in G1 phase. Nucleus 2, 146–157 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to F. Martino and D. Rhodes for help in setting up NCP preparations, to A. F. Straight for providing plasmids for expression of CENP-A:H4, to K. A. Davey for plasmids to produce the ‘601’ 145-bp nucleosomal DNA, to C. Smith for setting up conditions for CENP-H depletion, to G. Ossolengo of the antibody facility at the European Institute of Oncology in Milan (Italy) for help with antibody production, and all other members of the Musacchio laboratory for discussions. A.C.F. is supported by an EMBO long-term fellowship (ALTF 1096-2012) and Marie Curie Intra-European Fellowship. A.M. acknowledges funding by the European Union’s 7th Framework Program Integrated Project MitoSys, the Horizon 2020 ERC agreement RECEPIANCE, and the DFG’s Collaborative Research Centre 1093. F.H. is supported by the European Research Council (MolStruKT StG number 638218) and by an LMU excellent junior grant.

Author information

Authors and Affiliations

Authors

Contributions

J.R.W., K.K., A.C.F. and A.M. designed the experiments. J.R.W, K.K., A.C.F., F.B., J.K., A.P., S.W., M.P. and S.P. purified proteins. D.V. and F.B. purified nucleosomes. D.P. created the engineered nucleosomes used in AUC experiments. J.R.W. performed gel filtration experiments. A.P. performed AUC experiments. K.K. performed cell biology experiments. A.C.F. performed microtubule binding experiments. J.F. and F.H. performed crosslinking and mass-spec experiments. A.M. coordinated the working team. J.R.W. and A.M. wrote the paper.

Corresponding authors

Correspondence to John R. Weir or Andrea Musacchio.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information

Nature thanks A. Desai and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Building blocks of the kinetochore.

Schematic organization of protein and subcomplexes used in this study, with essential structural features. CENP-A is a histone H3 variant. Crucial to its function in kinetochore assembly are the so-called CATD box and the C-terminal region, which are believed to interact with CENP-N and CENP-C, respectively10,11,39. For our reconstitution studies, we reconstituted human CENP-A:H4 tetramers and combined them with X. laevis H2A:H2B dimers. Nucleosome core particles containing histone H3 were reconstituted with X. laevis H3, H4, H2A, and H2B (see Methods). CENP-C can be thought of as a blueprint for kinetochore assembly, with binding motifs for outer and inner kinetochore subunits ordered from the N to the C terminus. The N-terminal region starts with a binding site for the Mis12 complex40,41, followed by a binding site for the CENP-HIKM complex19. Two related nucleosome-binding motifs have been identified, in the so-called ‘central region’ and ‘CENP-C motif’11. The nucleosome-binding motifs interact with the H2A:H2B dimer and with the C-terminal region of CENP-A11. Finally, the dimerization motif has a cupin-like fold42. The C-terminal region also binds to M18BP1 (refs 43, 44), which is involved in CENP-A deposition. The two subunits of the CENP-LN complex have similar size and are structurally related, as revealed by the crystal structure of their S. cerevisiae homologues15. The four-subunit CENP-HIKM complex contains a tight subcomplex of the CENP-H and CENP-K subunits19. CENP-M is a pseudo-Ras-like small GTPase that has lost the ability to bind GTP19. It interacts with CENP-I and is required for its stability19, but no CENP-M orthologue has been identified in S. cerevisiae, whereas Ctf3 is the CENP-I orthologue in this organism (see Fig. 4a). Structurally, CENP-I may resemble the HEAT-repeat α-solenoid structure of Importin-β (ref. 19). The four-subunit NDC80 complex is crucial for microtubule-binding by kinetochores7,8. It is a dumbbell-shaped, elongated protein with large coiled-coil domains23,45. Calponin-homology (CH) domains near the N terminus of the NDC80 and NUF2 subunits have been implicated in microtubule-binding23,45. The RWD domains of the SPC24 and SPC25 subunits target the NDC80 complex to the kinetochore46,47 through interactions with the MIS12 complex. The four-subunit MIS12 complex remains structural uncharacterized, except for low-resolution negative-stain electron microscopy analyses47,48,49,50. It is a hub of interactions, including interactions with the CENP-C complex (discussed above), the NDC80 complex (also discussed above), and the Knl1 subunit of the Knl1 complex49. The two-subunit KNL1 complex plays a crucial role in spindle assembly checkpoint signalling51. The C-terminal region of KNL1, the largest known core kinetochore subunits, consists of tandem RWD domains and is sufficient with an interaction with the MIS12 complex47,49. A longer region, comprising approximately the last 300 residues, is also sufficient for tight binding to ZWINT. For our studies, we used a construct encompassing residues KNL12000–2311 that was endowed with the ability to bind the MIS12 complex and ZWINT.

Extended Data Figure 2 SEC analyses.

The indicated samples (at a concentration of 10 μM, 5 μM for nucleosome core particles) were loaded on the indicated SEC column and the resulting elution fractions were analysed by SDS–PAGE. a, CENP-LN complex. Note that CENP-L and CENP-N migrate identically in these gels because of their almost identical mass. They can be distinguished by selective addition of a tag, as shown in Fig. 1c. b, CENP-C1–544 complex. c, CENP-HIKM. d, KMN network. e, CENP-ANCP; the lower panel is a MidoriGreen-stained agarose gel of the same fractions analysed by SDS–PAGE in the upper panel. f, H3NCP; bottom panel as in e.

Extended Data Figure 3 Additional CENP-A binding experiments.

a, EMSAs were used to assess relative binding affinity of H3 or CENP-A NCPs to the CENP-LN complex. Quantification of binding data predicts the indicated dissociation constants. In quantifications of ac, the mean ± s.d. from three independent experiments is shown. b, CENP:CHIKMLN was titrated against Alexa-647-labelled CENP-A NCPs (purple trace) or H3 NCPs (grey trace) in an EMSA assay. Experimental triplicates were performed, and the approximate dissociation constant determined. CENP:CHIKMLN binds with approximately sevenfold higher affinity to CENP-A NCPs than to H3 NCPs. c, EMSA assays were performed using Alexa-647-labelled DNA in free form (black trace), in complex with H3 containing octamers (grey trace), or in complex with CENP-A containing octamers (purple trace). CENP-HIKM complex was titrated against the DNA or NCPs. No binding preference emerged. d, Biotinylated CENP-A NCPs were used as bait to pull down CENP:CHIKMLN complex. Interactions were then competed for using an increasing concentration of free (non-biotylated) CENP-A NCPs (left) or H3 NCPs (right). The ratio of CENP-I to CENP-A was plotted in the lower graph. Free CENP-A NCPs effectively compete off biotinylated CENP-A NCPs from the CHIKMLN complex. Free H3 NCPs are unable to do so, even at concentrations 20-fold the biotinylated bait. The assay was performed in 200 mM NaCl, and used a shorter construct of CENP-C (189–544) owing to the greater stability of this construct at lower salt concentrations and to better separation of the CENP-C and CENP-I bands on SDS–PAGE for analysis. e, Biotinylated nucleosomes were used as bait to pull down CENP:CHIKMLN complex. Pull-downs were performed at increasing salt concentrations from 100–300 mM NaCl. CENP-A nucleosomes maintained a strong interaction with CENP:CHIKMLN in 300 mM salt. H3 NCPs lost the interaction with CENP:CHIKMLN at NaCl concentrations above 200 mM.

Extended Data Figure 4 Binding assays and analytical ultracentrifugation.

a, Normalized sedimentation coefficient (c(s)) distributions of the respective sedimentation velocity runs. The data were collected at 280 nm and the size distribution analysis of the sedimentation coefficient was performed with SEDFIT38 software using a continuous c(s) model. The rotor was spun at 42,000 rpm and equilibrated at 20 °C for 1 h before the start of the run. b, Normalized c(s) distributions of the indicated sedimentation velocity runs. The data were collected at 497 nm (thus analysing signals from CENP-HI57-CKM complex labelled with Alexa Fluor 488) and the size distribution analysis of the sedimentation coefficient was performed with SEDFIT using a continuous c(s) model. The rotor was spun at 42,000 rpm and equilibrated at 10 °C for 2 h before the start of the run. We were unable to carry out runs with isolated CENP-C1–544, CENP-C189–544, or CENP-LN complex, owing to sample instability during the centrifugation experiments. c, Normalized c(s) distributions of the indicated sedimentation velocity runs. The data were collected at 401 nm to monitor sedimentation of blue fluorescent protein (BFP) in chimaeric nucleosomes consisting of residues 2–75 of histone H3.1 and residues 75–140 of CENP-A (see Methods). The chimaeric nucleosomes were mixed with a threefold excess of CHIKMLN complex (containing CENP-C189–544: that is, a construct devoid of the binding domain for the MIS12 complex) to saturate binding.

Extended Data Figure 5 Kinetochore localization studies.

a, Representative images showing kinetochore levels in interphase cells of CENP-A, CENP-C, and CENP-HK (with an antibody raised against the CENP-HK complex) in Flp-In T-REx HeLa cells upon RNAi-based depletion of the indicated proteins. Kinetochores were visualized with anti-CENP-A sera. Scale bar, 10 μm. Magnification 630×. b, Western blots documenting protein depletion. RNAi-based depletion of CENP-C appears incomplete by western blotting, whereas it appears to be very penetrant in immunofluorescence experiments. We have described this phenomenon before20, and found that decreased CENP-C silencing correlates with the higher degree of cell confluence (~80%) for the relatively large-scale RNAi preparations required for western blotting compared with immunofluorescence (where we start with cells at ~30% confluence).

Extended Data Figure 6 Incorporation of CENP-TWSX.

In an in vitro pull-down assay, CENP-ANCP reconstituted with biotinylated DNA were incubated with streptavidin-coated beads and the other recombinant kinetochore species indicated in the INPUT (top) panel of the figure. Beads were recovered by centrifugation and washed, and proteins bound to the solid phase (PULLDOWN, bottom) were visualized by SDS–PAGE followed by Coomassie blue staining. Binding of CENP-TWSX tetramer (which contains only the histone fold domain of CENP-T) was contingent to binding of CENP-C1–544 and CENP-HI57-CKM.

Extended Data Figure 7 Topology of the kinetochore.

Using XL–MS, the inter-peptide interactions within the kinetochore sample were analysed. Intra-protein crosslinks are shown in red, intra-subcomplex crosslinks are shown in orange, inter-subcomplex crosslinks in black. Proteins are coloured according to their subcomplex: CENP-ANCP, purple; CENP-C1–544, red; CENP-HIKM, green; CENP-LN, blue; MIS12-C, peach; KNL1-C, orange; NDC80-C, yellow.

Extended Data Figure 8 Microtubule-binding experiments.

a, Description of reagents and buffer used in experiments in b and in Fig. 3b. b, Rhodamine-labelled microtubules (red channel) were tethered to glass coverslips and incubated in the presence of GFP–KMN (green), Alexa-405-labelled CHIKMLN (blue), or Alexa-647-labelled CENP-ANCP or H3NCP (purple), and combinations thereof. Only CENP-ANCP translocated to microtubules, whereas H3NCP did not. Single microtubules from these images have already been shown in Fig. 3b. c, Quantification of experiments, already shown in Fig. 3b, c. Shown for each channel is mean ± s.e.m. from at least 20 microtubules in at least 2 independent experiments.

Extended Data Figure 9 CENP-C545–943 does not interact with CCAN subunits.

a, SEC analysis of CENP-ANCP (purple), CENP-C631–943 (red trace), and their combination (green trace) shows a stoichiometric interaction. b, SEC analysis of CENP-LN (blue trace), CENP-HI57-CKM (green trace), CENP-C545–943 (red trace), and their combination (orange trace). No apparent shift of CENP-C545–943 was observed. c, Summary of known interactions at the centromere–kinetochore interface. The N-terminal region of CENP-C (exemplified by CENP-C1–544) binds the KMN, the CHIKMLN, and a CENP-A nucleosome. The C-terminal region of CENP-C (exemplified by CENP-C545–943) does not bind core kinetochore components (this study) but interacts with CENP-A loading machinery, including the Mis18 complex, which in turn recruits the CENP-A chaperone HJURP52,53. Each half of CENP-C contains a nucleosome-binding motif, and has therefore the potential to interact with two adjacent nucleosomes. After DNA replication, when CENP-A levels are halved, CENP-A is replaced with H3 (H3.3, refs 54, 55). After mitosis, the C-terminal region of CENP-C contributes to recruit machinery that replaces H3 with CENP-A.

Supplementary information

Supplementary Data

This file contains Supplementary Table 1 (XLSX 62 kb)

Supplementary Data

This file contains Supplementary Table 2 (XLSX 89 kb)

Supplementary Information

This file contains the original scanned gels before cropping, with an indication of the cropped area as well as reference to the original figure number in the main body of the paper or in the Extended Data Figures. (PDF 2136 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weir, J., Faesen, A., Klare, K. et al. Insights from biochemical reconstitution into the architecture of human kinetochores. Nature 537, 249–253 (2016). https://doi.org/10.1038/nature19333

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature19333

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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