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:

Parental histone transfer caught at the replication fork

Abstract

In eukaryotes, DNA compacts into chromatin through nucleosomes1,2. Replication of the eukaryotic genome must be coupled to the transmission of the epigenome encoded in the chromatin3,4. Here we report cryo-electron microscopy structures of yeast (Saccharomyces cerevisiae) replisomes associated with the FACT (facilitates chromatin transactions) complex (comprising Spt16 and Pob3) and an evicted histone hexamer. In these structures, FACT is positioned at the front end of the replisome by engaging with the parental DNA duplex to capture the histones through the middle domain and the acidic carboxyl-terminal domain of Spt16. The H2A–H2B dimer chaperoned by the carboxyl-terminal domain of Spt16 is stably tethered to the H3–H4 tetramer, while the vacant H2A–H2B site is occupied by the histone-binding domain of Mcm2. The Mcm2 histone-binding domain wraps around the DNA-binding surface of one H3–H4 dimer and extends across the tetramerization interface of the H3–H4 tetramer to the binding site of Spt16 middle domain before becoming disordered. This arrangement leaves the remaining DNA-binding surface of the other H3–H4 dimer exposed to additional interactions for further processing. The Mcm2 histone-binding domain and its downstream linker region are nested on top of Tof1, relocating the parental histones to the replisome front for transfer to the newly synthesized lagging-strand DNA. Our findings offer crucial structural insights into the mechanism of replication-coupled histone recycling for maintaining epigenetic inheritance.

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: The overall structure of an endogenous replisome.
Fig. 2: The evicted histone hexamer and its chaperones at the replication fork.
Fig. 3: Deposition of the histone hexamer by Mcm2 NTE onto the top surface of Tof1.
Fig. 4: The detailed interactions between the Mcm2 NTE and Tof1.
Fig. 5: eSPAN analyses of parental histone recycling mediated by the interaction between Tof1 and the Mcm2 NTE.
Fig. 6: Model of parental histone recycling to the lagging strand mediated by the replisome and FACT at the replication fork.

Similar content being viewed by others

Data availability

Coordinates and cryo-EM map data have been deposited at the Protein Data Bank and Electron Microscopy Data Bank under accession codes EMD-38317 and PDB 8XGC (composite map and the global atomic model), EMD-38316 (global structure, conformation-1), EMD-38314 (global structure, conformation-2), EMD-38313 (optimized local density map of Spt16-MD–histone) and EMD-38315 (optimized local density map of Polε). The raw sequencing data reported in this paper have been deposited in the Genome Sequence Archive at the National Genomics Data Center, Beijing Institute of Genomics (China National Center for Bioinformation), Chinese Academy of Sciences, under accession number GSA CRA012495. The data are publicly accessible online (https://bigd.big.ac.cn/gsa).

References

  1. Zhou, K., Gaullier, G. & Luger, K. Nucleosome structure and dynamics are coming of age. Nat. Struct. Mol. Biol. 26, 3–13 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Sekine, S. I., Ehara, H., Kujirai, T. & Kurumizaka, H. Structural perspectives on transcription in chromatin. Trends Cell Biol. https://doi.org/10.1016/j.tcb.2023.07.011 (2023).

  3. Yung, P. Y. K. & Elsasser, S. J. Evolution of epigenetic chromatin states. Curr. Opin. Chem. Biol. 41, 36–42 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Du, W. et al. Mechanisms of chromatin-based epigenetic inheritance. Sci. China Life Sci. 65, 2162–2190 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Stewart-Morgan, K. R., Petryk, N. & Groth, A. Chromatin replication and epigenetic cell memory. Nat. Cell Biol. 22, 361–371 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Zhang, W., Feng, J. & Li, Q. The replisome guides nucleosome assembly during DNA replication. Cell Biosci. 10, 37 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shan, Z. et al. The patterns and participants of parental histone recycling during DNA replication in Saccharomyces cerevisiae. Sci. China Life Sci. 66, 1600–1614 (2023).

    Article  CAS  PubMed  Google Scholar 

  8. Attali, I., Botchan, M. R. & Berger, J. M. Structural mechanisms for replicating DNA in eukaryotes. Annu. Rev. Biochem. 90, 77–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Bell, S. P. & Labib, K. Chromosome duplication in Saccharomyces cerevisiae. Genetics 203, 1027–1067 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yao, N. Y. & O’Donnell, M. E. The DNA replication machine: structure and dynamic function. Subcell. Biochem. 96, 233–258 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Gao, Y. & Yang, W. Different mechanisms for translocation by monomeric and hexameric helicases. Curr. Opin. Struct. Biol. 61, 25–32 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Yeeles, J. T. P., Janska, A., Early, A. & Diffley, J. F. X. How the eukaryotic replisome achieves rapid and efficient DNA replication. Mol. Cell 65, 105–116 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kurat, C. F., Yeeles, J. T. P., Patel, H., Early, A. & Diffley, J. F. X. Chromatin controls DNA replication origin selection, lagging-strand synthesis, and replication fork rates. Mol. Cell 65, 117–130 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yeeles, J. T., Deegan, T. D., Janska, A., Early, A. & Diffley, J. F. Regulated eukaryotic DNA replication origin firing with purified proteins. Nature 519, 431–435 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  15. Formosa, T. The role of FACT in making and breaking nucleosomes. Biochim. Biophys. Acta 1819, 247–255 (2013).

    Article  PubMed  Google Scholar 

  16. Safaric, B. et al. The fork protection complex recruits FACT to reorganize nucleosomes during replication. Nucleic Acids Res. 50, 1317–1334 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ehara, H., Kujirai, T., Shirouzu, M., Kurumizaka, H. & Sekine, S. I. Structural basis of nucleosome disassembly and reassembly by RNAPII elongation complex with FACT. Science 377, eabp9466 (2022).

    Article  CAS  PubMed  Google Scholar 

  18. Farnung, L., Ochmann, M., Engeholm, M. & Cramer, P. Structural basis of nucleosome transcription mediated by Chd1 and FACT. Nat. Struct. Mol. Biol. 28, 382–387 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu, Y. et al. FACT caught in the act of manipulating the nucleosome. Nature 577, 426–431 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Georgescu, R. et al. Structure of eukaryotic CMG helicase at a replication fork and implications to replisome architecture and origin initiation. Proc. Natl Acad. Sci. USA 114, E697–E706 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Douglas, M. E., Ali, F. A., Costa, A. & Diffley, J. F. X. The mechanism of eukaryotic CMG helicase activation. Nature 555, 265–268 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun, J. et al. The architecture of a eukaryotic replisome. Nat. Struct. Mol. Biol. 22, 976–982 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Baretić, D. et al. Cryo-EM structure of the fork protection complex bound to CMG at a replication fork. Mol Cell 78, 926–940 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jones, M. L., Baris, Y., Taylor, M. R. G. & Yeeles, J. T. P. Structure of a human replisome shows the organisation and interactions of a DNA replication machine. EMBO J. 40, e108819 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rzechorzek, N. J., Hardwick, S. W., Jatikusumo, V. A., Chirgadze, D. Y. & Pellegrini, L. CryoEM structures of human CMG-ATPγS-DNA and CMG-AND-1 complexes. Nucleic Acids Res. 48, 6980–6995 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xu, Z. et al. Synergism between CMG helicase and leading strand DNA polymerase at replication fork. Nat. Commun. 14, 5849 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Huang, H. et al. A unique binding mode enables MCM2 to chaperone histones H3-H4 at replication forks. Nat. Struct. Mol. Biol. 22, 618–626 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, H., Wang, M., Yang, N. & Xu, R. M. Structure of the quaternary complex of histone H3-H4 heterodimer with chaperone ASF1 and the replicative helicase subunit MCM2. Protein Cell 6, 693–697 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Evrin, C., Maman, J. D., Diamante, A., Pellegrini, L. & Labib, K. Histone H2A-H2B binding by Pol α in the eukaryotic replisome contributes to the maintenance of repressive chromatin. EMBO J. https://doi.org/10.15252/embj.201899021 (2018).

  30. Li, Z. et al. DNA polymerase α interacts with H3-H4 and facilitates the transfer of parental histones to lagging strands. Sci. Adv. 6, eabb5820 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Petryk, N. et al. MCM2 promotes symmetric inheritance of modified histones during DNA replication. Science 361, 1389–1392 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Gan, H. et al. The Mcm2-Ctf4-Polα axis facilitates parental histone H3-H4 transfer to lagging strands. Mol. Cell 72, 140–151 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yu, C. et al. A mechanism for preventing asymmetric histone segregation onto replicating DNA strands. Science 361, 1386–1389 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Flury, V. et al. Recycling of modified H2A-H2B provides short-term memory of chromatin states. Cell 186, 1050–1065 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Jenkyn-Bedford, M. et al. A conserved mechanism for regulating replisome disassembly in eukaryotes. Nature 600, 743–747 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Tsunaka, Y., Fujiwara, Y., Oyama, T., Hirose, S. & Morikawa, K. Integrated molecular mechanism directing nucleosome reorganization by human FACT. Genes Dev. 30, 673–686 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jones, M. L., Aria, V., Baris, Y. & Yeeles, J. T. P. How Pol α-primase is targeted to replisomes to prime eukaryotic DNA replication. Mol. Cell 83, 2911–2924 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yu, C. et al. Strand-specific analysis shows protein binding at replication forks and PCNA unloading from lagging strands when forks stall. Mol. Cell 56, 551–563 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xu, M. et al. Partitioning of histone H3-H4 tetramers during DNA replication-dependent chromatin assembly. Science 328, 94–98 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Groth, A. et al. Regulation of replication fork progression through histone supply and demand. Science 318, 1928–1931 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  41. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife https://doi.org/10.7554/eLife.42166 (2018).

  42. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Rohou, A. & Grigorieff, N. CTFFIND4: Fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Bepler, T. et al. Positive-unlabeled convolutional neural networks for particle picking in cryo-electron micrographs. Nat. Methods 16, 1153–1160 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bepler, T., Kelley, K., Noble, A. J. & Berger, B. Topaz-Denoise: general deep denoising models for cryoEM and cryoET. Nat. Commun. 11, 5208 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, educators, and developers. Protein Sci. 30, 70–82 (2021).

    Article  CAS  PubMed  Google Scholar 

  49. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  50. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  ADS  CAS  PubMed  Google Scholar 

  52. Li, Q. et al. Acetylation of histone H3 lysine 56 regulates replication-coupled nucleosome assembly. Cell 134, 244–255 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Chen, K. et al. DANPOS: dynamic analysis of nucleosome position and occupancy by sequencing. Genome Res. 23, 341–351 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Labib and T. Formosa for antibodies; and the members of the Biological Cryo-EM Center at the Hong Kong University of Science and Technology (HKUST) for data collection. The Cryo-EM Center at HKUST is supported by a donation from the Lo Kwee Seong Foundation. The computation was supported by the High-Performance Computing Platform of Peking University (PKU). We also thank the cryo-EM platform of PKU and the National Center for Protein Sciences at PKU for technical assistance. The work was supported by the National Natural Science Foundation of China (32321163647 to N.G., 31830048 to Q.L. and 31922036 to N.L.), the National Key R&D Program of China (2019YFA0508900 to Q.L. and 2019YFA0508904 to N.G.), the Beijing Outstanding Young Scientist Program (BJJWZYJH01201910001005) to Q.L., and the Research Grants Council (RGC) of Hong Kong (GRF17119022, GRF17109623, C7009-20GF, and CRS_HKU705/23 to Y. Zhai).

Author information

Authors and Affiliations

Authors

Contributions

Y. Zhai, N.G. and Q.L. conceived and supervised the project. Y.G., J. Li, Z.X. and Y. Zhai purified the replisome samples and conducted biochemical assays. J. Lin and X.D.L. performed quantitative MS analysis. D.Y. and Y.G. prepared cryo samples. D.Y., Yingyi Zhang and S.D. collected cryo-EM datasets. N.L. processed images. N.G., N.L. and Y. Zhai built atomic models. Yujie Zhang, J.F. and Q.L. performed eSPAN analyses. Y. Zhai, N.G., N.L., Y.G., Yujie Zhang, Q.L., J. Lin, X.D.L., B.K.T., Y.L. and K.Z. analysed the data, prepared the figures and wrote the manuscript.

Corresponding authors

Correspondence to Bik Kwoon Tye, Qing Li, Ning Gao or Yuanliang Zhai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Hitoshi Kurumizaka 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 figures and tables

Extended Data Fig. 1 Purification of the endogenous replisome.

a, A flowchart of the procedures for replisome purification from early S chromatin of the yeast strain PSF2-3xFlag. b-c, SDS-PAGE analysis of the glycerol gradient (20-40%) fractions of the replisome sample eluted from anti-Flag affinity purification. The fractions were resolved and visualized by silver staining (b) and immunoblotting of the proteins as indicated (c). Fractions 5-8 containing replisome-FACT-histones were pooled and processed for mass spectrometry analysis. Based on this result, similar fractions containing crosslinked samples after grafix were pooled and processed for further EM analyses. Similar results were obtained at least in two independent experiments. d, The major proteins associated with the endogenous replisome identified by quantitative liquid chromatography mass spectrometry (LC-MS). For gel source data, see Supplementary Fig. 1.

Extended Data Fig. 2 Cryo-EM image processing of the replisome sample.

a, A representative raw cryo-EM image of the replisome sample. b-c, 2D class averages of negative staining (b) and cryo-EM (c) replisome particles. d-i, Workflow of image processing of the replisome cryo-EM images. See Methods for details. j, Corrected (the right line group) and phase randomized (the left line group) FSC curves of the final global and local density maps for the replisome complex. k, The local resolutions of the composite replisome map were colour coded. l-q, Cryo-EM densities for indicated regions of the replisome.

Extended Data Fig. 3 Conformational change in FACT during substrate transaction.

a, c, Different views of the local density map of the FACT-histones complex bound with parental duplex DNA. b, d, same as (a, c) respectively but superimposed with the atomic model. e-f, FACT-DNA interaction. Comparison of the Spt16-DD/Pob3-DD-DNA structure from the replisome (this study) with those from FACT-hexasome (PDB: 6UPK) (e) and RNA Polymerase II-Spt4/5-nucleosome-FACT (PDB: 7NKY) (f). Spt16-DD/Pob3-DD was used as a reference for alignment. g-j, Comparison of the FACT structures from the replisome-histone hexamer-FACT (this study) (g), human FACT-hexasome (PDB: 6UPK) (i), yeast RNA Polymerase II-Spt4/5-nucleosome-FACT (PDB: 7NKY) (j), and RNA polymerase II elongation complex-nucleosome-FACT (Komagataella phaffii) (EC58hex, PDB: 7XTI) (j). Spt16-DD/Pob3-DD was used as a reference for alignment. k-m, Superimposition of the FACT structure from the replisome (this study, g) with those from h-j to highlight the conformational changes in Spt16 bound with different substrates as indicated, respectively. Spt16-DD/Pob3-DD was used as a reference for alignment. n-w, The relevant structures from g-m were aligned using histones as a reference to illustrate the conformational changes in both Spt16 and Pob3 while relocating histones onto Tof1 at replication fork during parental histone recycling. The histones and DNA from the replisome were omitted to highlight the movement of Spt16-MD in r-w.

Extended Data Fig. 4 The histone hexamer is shielded by its chaperones at the replication fork.

a-d, Schematic illustration of the histone-chaperone interaction in the replisome. e-f, Side views of the electrostatic surfaces of the histone hexamer highlighting its associations with Spt16-MD, Mcm2-NTE, and Tof1-HTH for shielding its DNA binding surfaces. i-l, same as (e-f) but superimposed with the nucleosomal DNA from an intact nucleosome (PDB: 1ID3).

Extended Data Fig. 5 Small displacement of H2A-H2Bproximal by Spt16-MD.

a-c, Comparison of the histone structures from the replisome (a) and an intact nucleosome (PDB: 1ID3) (b). Superimposition of these two structures using (H3-H4)2 as a reference for alignment (c). H3-αN and H2A-docking segment were coated with transparent surface presentation to highlight conformational changes occurring upon these regions in the replisome. d, Same as (c) but with Spt16-MD from the replisome visible to illustrate the steric conflict between Spt16-MD and H3-αN/H2A-docking segment on H3-H4 tetramer. e, Zoomed-in view of the boxed region (red box) in (d) but with the region from the replisome shown only. f, Same as (e) but shown with the H3 from the intact nucleosome (grey) and with Spt16-MD from the replisome superimposed. g, Zoomed-in view of the boxed region (green box) in (d) but with the region from the replisome shown only. The key residues involved in the interaction between Spt16-MD and H2A-docking segment are shown in stick and labelled. h, Same as (g) but shown with the H2A-docking segment from the intact nucleosome (red) and with Spt16-MD from the replisome superimposed. i, Same as (d) but with a 90°-rotation. The (H3-H4)2 tetramer is not shown to highlight a 13°-rotation in the H2A-H2Bproximal upon Spt16-MD binding.

Extended Data Fig. 6 The arrangement of Mcm2-NTE around the evicted histones is pre-determined by Spt16-MD and H2A-H2Bproximal.

a, Structural comparison of the (H3-H4)2/MCM2-NTE interactions from the structures of human (H3-H4)2-MCM2-HBD (PDB:5BNV) and yeast histone hexamer-Mcm2-NTE-Spt16-MD from the replisome (this study). H3-H4 was used as a reference for alignment. h: human; y: yeast; M2: MCM2. b, Same as (a) but with hM2-NTE-1 and yM2-NTE shown only. c, Same as (a) but with y(H3-H4)2 shown in surface presentation and other subunits and motifs in cartoon presentation. d-e, Zoomed-in views of the boxed regions in (c).

Extended Data Fig. 7 eSPAN analysis of the interaction between Tof1 and Mcm2.

a, Snapshot of MNase-seq, MNase-BrdU-IP-seq, H3K4me3-ChIP-seq, H3K4me3-eSPAN, H3K56ac-ChIP-seq, and H3K56ac-eSPAN datasets around replication origins ARS1309 and ARS1310 in wild-type (WT), tof1-3A, tof1ΔHTH, mcm2-6A, and mcm2ΔL. The scale bar represents a 10-kilo base pair (kbp) DNA region. b, DNA content analysis of the eSPAN samples. Cellular DNA content was measured by flow cytometry with PI staining. Similar results were obtained in two independent experiments. See Methods for more details.

Extended Data Fig. 8 The interaction between Tof1 and Mcm2 affects the relative amounts of the newly synthesized histone deposition between the leading and lagging strands.

a, Line plots of the H3K56ac eSPAN bias show the relative amount of new histone deposition between the two daughter strands in wild-type (WT), tof1-3A, and tof1ΔHTH cells. The H3K56ac-eSPAN bias around 139 early replication origins of the autonomous replication sequences (ACSs) was calculated. b, Box plots of the H3K56ac-eSPAN bias around the 139 ACSs in WT, tof1-3A, and tof1ΔHTH mutant cells are shown (n = 139). c, Box plots showing the H3K56ac-eSPAN density on both leading and lagging strands in WT, tof1-3A, and tof1ΔHTH cells (n = 139). d, Line plots of the H3K56ac eSPAN bias around the 139 ACSs in wild-type (WT), mcm2-6A, and mcm2ΔL cells are shown. e, Box plots of the H3K56ac-eSPAN bias around the 139 ACSs in WT and mcm2-6A or mcm2ΔL mutant cells are shown (n = 139). f, Box plots showing the H3K56ac-eSPAN density on both leading and lagging strands in WT, mcm2-6A, and mcm2ΔL cells (n = 139). g-h, Heatmap analysis of the H3K56ac-eSPAN bias around 139 ACSs in WT, tof1-3A, tof1ΔHTH (g), mcm2-6A, and mcm2ΔL (h) cells. Box plots (b, c, e, f) show the median, minimal, maximal, and 25% and 75% quartiles values. P values calculated using two-sided rank-sum Wilcoxon test. Similar results were obtained in two independent experiments. See Methods for more details.

Extended Data Fig. 9 The asymmetric histone partitioning between daughter strands in the disrupted Tof1-Mcm2 interaction cells does not result from defects in DNA replication.

a-d, The statistical results of correlation between BrdU incorporation level and H3K4me3-eSPAN value at each ACS region in WT (a), tof1-3A (b), mcm2-6A (c) and mcm2ΔL (d) cells. Dot scatterplot showing the distribution of the H3K4me3-eSPAN bias at early replication origins (n = 139). e-h, The statistical results of correlation between BrdU incorporation level and H3K56ac-eSPAN value at each ACS region in WT (e), tof1-3A (f), mcm2-6A (g) and mcm2ΔL (h) cells. Dot scatterplot showing the distribution of the H3K56ac-eSPAN bias at early replication origins (n = 139). Similar results were obtained in two independent experiments.

Extended Data Table 1 Cryo-EM data collection, refinement, and validation statistics

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Supplementary Tables 2 and 3.

Reporting Summary

Peer Review File

Supplementary Table 1

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

Li, N., Gao, Y., Zhang, Y. et al. Parental histone transfer caught at the replication fork. Nature 627, 890–897 (2024). https://doi.org/10.1038/s41586-024-07152-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07152-2

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