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:

H4S47 O-GlcNAcylation regulates the activation of mammalian replication origins

Abstract

The transmission and maintenance of genetic information in eukaryotic cells relies on the faithful duplication of the entire genome. In each round of division, excessive replication origins are licensed, with only a fraction activated to give rise to bi-directional replication forks in the context of chromatin. However, it remains elusive how eukaryotic replication origins are selectively activated. Here we demonstrate that O-GlcNAc transferase (OGT) enhances replication initiation by catalyzing H4S47 O-GlcNAcylation. Mutation of H4S47 impairs DBF4-dependent protein kinase (DDK) recruitment on chromatin, causing reduced phosphorylation of the replicative helicase mini-chromosome maintenance (MCM) complex and compromised DNA unwinding. Our short nascent-strand sequencing results further confirm the importance of H4S47 O-GlcNAcylation in origin activation. We propose that H4S47 O-GlcNAcylation directs origin activation through facilitating MCM phosphorylation, and this may shed light on the control of replication efficiency by chromatin environment.

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: H4S47 O-GlcNAcylation is required for efficient DNA replication.
Fig. 2: OGT regulates DNA replication through H4S47 O-GlcNAcylation.
Fig. 3: H4S47 O-GlcNAcylation facilitates origin activation by regulating MCMs.
Fig. 4: H4S47 O-GlcNAcylation recruits DDK through direct interaction with DBF4.
Fig. 5: S47 O-GlcNAcylation orients MCM association with H4 on chromatin.
Fig. 6: H4S47 O-GlcNAcylation fosters DNA unwinding for origin activation.
Fig. 7: Genome-wide investigation of H4S47 O-GlcNAcylation function at origins.

Similar content being viewed by others

Data availability

All the genome-wide sequencing datasets generated and analyzed during the current study are submitted to Gene Expression Omnibus (GEO) repository under the GEO accession number GSE205673. Human genome (hg38) was accessed from https://hgdownload.soe.ucsc.edu/goldenPath/hg38. Mouse genome (mm10) was accessed from https://hgdownload.soe.ucsc.edu/goldenPath/mm10. The mixed hg38-mm10 genome was made by concatenated fasta files of human genome (hg38) and mouse genome (mm10), with chromosome names of mouse genome modified to have the prefix ‘mm10_’. The concatenated fasta file was used to build bowtie2 reference using bowtie-build63. The fasta index file was generated using samtools faidx68 and can be found at https://github.com/maximewen/custom_ref. Source data are provided with this paper.

Code availability

The custom python script used for filtering pair-end reads can be found at https://github.com/maximewen/DNA_replication.

References

  1. Fragkos, M., Ganier, O., Coulombe, P. & Mechali, M. DNA replication origin activation in space and time. Nat. Rev. Mol. Cell Biol. 16, 360–374 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Prioleau, M. N. & MacAlpine, D. M. DNA replication origins—where do we begin? Genes Dev. 30, 1683–1697 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Abbas, T., Keaton, M. A. & Dutta, A. Genomic instability in cancer. Cold Spring Harb. Perspect. Biol. 5, a012914 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Alabert, C. & Groth, A. Chromatin replication and epigenome maintenance. Nat. Rev. Mol. Cell Biol. 13, 153–167 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Blow, J. J., Ge, X. Q. & Jackson, D. A. How dormant origins promote complete genome replication. Trends Biochem. Sci. 36, 405–414 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Remus, D. & Diffley, J. F. Eukaryotic DNA replication control: lock and load, then fire. Curr. Opin. Cell Biol. 21, 771–777 (2009).

    Article  CAS  PubMed  Google Scholar 

  7. Im, J. S. et al. RecQL4 is required for the association of Mcm10 and Ctf4 with replication origins in human cells. Cell Cycle 14, 1001–1009 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kumagai, A., Shevchenko, A., Shevchenko, A. & Dunphy, W. G. Direct regulation of Treslin by cyclin-dependent kinase is essential for the onset of DNA replication. J. Cell Biol. 193, 995–1007 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. MacAlpine, D. M. & Almouzni, G. Chromatin and DNA replication. Cold Spring Harb. Perspect. Biol. 5, a010207 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Gilbert, D. M. In search of the holy replicator. Nat. Rev. Mol. Cell Biol. 5, 848–855 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Akerman, I. et al. A predictable conserved DNA base composition signature defines human core DNA replication origins. Nat. Commun. 11, 4826 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Margueron, R. & Reinberg, D. Chromatin structure and the inheritance of epigenetic information. Nat. Rev. Genet. 11, 285–296 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Probst, A. V., Dunleavy, E. & Almouzni, G. Epigenetic inheritance during the cell cycle. Nat. Rev. Mol. Cell Biol. 10, 192–206 (2009).

    Article  CAS  PubMed  Google Scholar 

  14. Miotto, B. & Struhl, K. HBO1 histone acetylase activity is essential for DNA replication licensing and inhibited by Geminin. Mol. Cell 37, 57–66 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tardat, M. et al. The histone H4 Lys 20 methyltransferase PR-Set7 regulates replication origins in mammalian cells. Nat. Cell Biol. 12, 1086–1093 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Sakabe, K., Wang, Z. & Hart, G. W. Beta-N-acetylglucosamine (O-GlcNAc) is part of the histone code. Proc. Natl Acad. Sci. USA 107, 19915–19920 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hart, G. W. Nutrient regulation of signaling and transcription. J. Biol. Chem. 294, 2211–2231 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Levine, Z. G. & Walker, S. The biochemistry of O-GlcNAc transferase: which functions make it essential in mammalian cells? Annu. Rev. Biochem. 85, 631–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Yang, X. & Qian, K. Protein O-GlcNAcylation: emerging mechanisms and functions. Nat. Rev. Mol. Cell Biol. 18, 452–465 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, Y. et al. O-GlcNAcylation destabilizes the active tetrameric PKM2 to promote the Warburg effect. Proc. Natl Acad. Sci. USA 114, 13732–13737 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liu, S. et al. RPA binds histone H3-H4 and functions in DNA replication-coupled nucleosome assembly. Science 355, 415–420 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Kang, B. et al. Phosphorylation of H4 Ser 47 promotes HIRA-mediated nucleosome assembly. Genes Dev. 25, 1359–1364 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Toledo, L., Neelsen, K. J. & Lukas, J. Replication catastrophe: when a checkpoint fails because of exhaustion. Mol. Cell 66, 735–749 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Ortiz-Meoz, R. F. et al. A small molecule that inhibits OGT activity in cells. ACS Chem. Biol. 10, 1392–1397 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bai, G. et al. HLTF promotes fork reversal, limiting replication stress resistance and preventing multiple mechanisms of unrestrained DNA synthesis. Mol. Cell 78, 1237–1251(2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Schmid, J. A. et al. Histone ubiquitination by the DNA damage response is required for efficient DNA replication in unperturbed S phase. Mol. Cell 71, 897–910 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Alver, R. C., Chadha, G. S., Gillespie, P. J. & Blow, J. J. Reversal of DDK-mediated MCM phosphorylation by Rif1–PP1 regulates replication initiation and replisome stability independently of ATR/Chk1. Cell Rep. 18, 2508–2520 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Feng, Y. et al. BRPF3–HBO1 regulates replication origin activation and histone H3K14 acetylation. EMBO J. 35, 176–192 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Cayrou, C. et al. The chromatin environment shapes DNA replication origin organization and defines origin classes. Genome Res. 25, 1873–1885 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Long, H. et al. H2A.Z facilitates licensing and activation of early replication origins. Nature 577, 576–581 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Hulke, M. L., Massey, D. J. & Koren, A. Genomic methods for measuring DNA replication dynamics. Chromosome Res. 28, 49–67 (2020).

    Article  CAS  PubMed  Google Scholar 

  33. Rausch, C. et al. Developmental differences in genome replication program and origin activation. Nucleic Acids Res. 48, 12751–12777 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Langley, A. R., Gräf, S., Smith, J. C. & Krude, T. Genome-wide identification and characterisation of human DNA replication origins by initiation site sequencing (ini-seq). Nucleic Acids Res. 44, 10230–10247 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Picard, F. et al. The spatiotemporal program of DNA replication is associated with specific combinations of chromatin marks in human cells. PLoS Genet. 10, e1004282 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Tubbs, A. et al. Dual roles of poly(dA:dT) tracts in replication initiation and fork collapse. Cell 174, 1127–1142 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yamazaki, S. et al. Rif1 regulates the replication timing domains on the human genome. EMBO J. 31, 3667–3677 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Caballero, M. et al. Comprehensive analysis of DNA replication timing across 184 cell lines suggests a role for MCM10 in replication timing regulation. Hum. Mol. Genet. 31, 2899–2917 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fujiki, R. et al. GlcNAcylation of histone H2B facilitates its monoubiquitination. Nature 480, 557–560 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chen, Q. & Yu, X. OGT restrains the expansion of DNA damage signaling. Nucleic Acids Res. 44, 9266–9278 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhang, S., Roche, K., Nasheuer, H. P. & Lowndes, N. F. Modification of histones by sugar beta-N-acetylglucosamine (GlcNAc) occurs on multiple residues, including histone H3 serine 10, and is cell cycle-regulated. J. Biol. Chem. 286, 37483–37495 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lercher, L. et al. Generation of a synthetic GlcNAcylated nucleosome reveals regulation of stability by H2A-Thr101 GlcNAcylation. Nat. Commun. 6, 7978 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Cheng, L. et al. DDA1, a novel oncogene, promotes lung cancer progression through regulation of cell cycle. J. Cell. Mol. Med. 21, 1532–1544 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nabeel-Shah, S., Ashraf, K., Pearlman, R. E. & Fillingham, J. Molecular evolution of NASP and conserved histone H3/H4 transport pathway. BMC Evol. Biol. 14, 139 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hammond, C. M. et al. DNAJC9 integrates heat shock molecular chaperones into the histone chaperone network. Mol. Cell 81, 2533–2548 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Benedict, B. et al. The RECQL helicase prevents replication fork collapse during replication stress. Life Sci. Alliance 3, e202000668 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Thangavel, S. et al. Human RECQ1 and RECQ4 helicases play distinct roles in DNA replication initiation. Mol. Cell. Biol. 30, 1382–1396 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Watson, L. J. et al. Cardiomyocyte Ogt is essential for postnatal viability. Am. J. Physiol. Heart Circ. Physiol. 306, H142–H153 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Ruan, H. B. et al. O-GlcNAc transferase enables AgRP neurons to suppress browning of white fat. Cell 159, 306–317 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bester, A. C. et al. Nucleotide deficiency promotes genomic instability in early stages of cancer development. Cell 145, 435–446 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Cornacchia, D. et al. Mouse Rif1 is a key regulator of the replication-timing programme in mammalian cells. EMBO J. 31, 3678–3690 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Fernandez-Vidal, A. et al. A role for DNA polymerase θ in the timing of DNA replication. Nat. Commun. 5, 4285 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Dileep, V., Rivera-Mulia, J. C., Sima, J. & Gilbert, D. M. Large-scale chromatin structure–function relationships during the cell cycle and development: insights from replication timing. Cold Spring Harb. Symp. Quant. Biol. 80, 53–63 (2015).

    Article  PubMed  Google Scholar 

  54. Köhler, C. et al. Cdc45 is limiting for replication initiation in humans. Cell Cycle 15, 974–985 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Aparicio, T., Guillou, E., Coloma, J., Montoya, G. & Méndez, J. The human GINS complex associates with Cdc45 and MCM and is essential for DNA replication. Nucleic Acids Res. 37, 2087–2095 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Toledo, L. I. et al. ATR prohibits replication catastrophe by preventing global exhaustion of RPA. Cell 155, 1088–1103 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Nakamura, K. et al. Proteome dynamics at broken replication forks reveal a distinct ATM-directed repair response suppressing DNA double-strand break ubiquitination. Mol. Cell 81, 1084–1099 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. D’Amico, D. et al. The RNA-binding protein PUM2 impairs mitochondrial dynamics and mitophagy during aging. Mol. Cell 73, 775–787.e10 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Chen, W. et al. The neurogenic compound P7C3 regulates the aerobic glycolysis by targeting phosphoglycerate kinase 1 in glioma. Front. Oncol. 11, 644492 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Stopfer, L. E., Mesfin, J. M., Joughin, B. A., Lauffenburger, D. A. & White, F. M. Multiplexed relative and absolute quantitative immunopeptidomics reveals MHC I repertoire alterations induced by CDK4/6 inhibition. Nat. Commun. 11, 2760 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, X. et al. Nuclear PGK1 alleviates ADP-dependent inhibition of CDC7 to promote DNA replication. Mol. Cell 72, 650–660 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Martin, M. J. Ej Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  63. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  65. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Orlando, D. A. et al. Quantitative ChIP–seq normalization reveals global modulation of the epigenome. Cell Rep. 9, 1163–1170 (2014).

    Article  CAS  PubMed  Google Scholar 

  67. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Marchal, C. et al. Genome-wide analysis of replication timing by next-generation sequencing with E/L Repli-seq. Nat. Protoc. 13, 819–839 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ryba, T., Battaglia, D., Pope, B. D., Hiratani, I. & Gilbert, D. M. Genome-scale analysis of replication timing: from bench to bioinformatics. Nat. Protoc. 6, 870–895 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Saldanha, A. J. Java Treeview—extensible visualization of microarray data. Bioinformatics 20, 3246–3248 (2004).

    Article  CAS  PubMed  Google Scholar 

  72. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank W.G. Dunphy (Division of Biology and Biological Engineering, California Institute of Technology) and Y. Gu (School of Medicine and Pharmacy, Ocean University of China) for providing helpful reagents. We acknowledge S. Lee (College of Pharmacy, Taipei Medical University), C. Xu, and L. Gong (School of Life Sciences, Northeast Normal University) for technical advice. This work was supported by grants from the National Natural Science Foundation of China (31600645 and 32070758 to Y.F., 31870896 to W.M., 32000412 to H.L., and 32000423 to Z.W). This work was also supported by the Ministry of Science and Technology of China (2022YFA1302801 to H.L.).

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and J.P. conducted the majority of the experiments and analyzed the data. H.L., L.L., and Z.W. carried out SNS-seq, Repli-seq and associated data analysis. K.D. performed immunofluorescence. J.P. and G.Z. assisted with chromatin immunoprecipitation. T.W. and L.Z. performed the DNA fiber assay. M.L. constructed Flag-H4WT, Flag-H4S47A, Flag-OGT, and GFP-OGT plasmids. Z.Z. analyzed mass spectrometry data. X.J. and Y.W. provided support in flow cytometry. Y.Z., M.W., and Y.F. wrote the paper. Y.F. supervised the project. All authors discussed the results and commented on the paper.

Corresponding author

Correspondence to Yunpeng Feng.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks John Hanover and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Beth Moorefield, Carolina Perdigoto and Dimitris Typas, in collaboration with the Nature Structural & Molecular Biology team. 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 H4S47 O-GlcNAcylation regulates DNA replication.

a, Detection of H4S47 O-GlcNAcylation by mass spectrometry. Histone H4 extracted from HEK293T cells were analyzed by mass spectrometry. Tandem mass spectrum of peptides with O-GlcNAcylation on S47 of H4 is shown. The y and b fragmentations were used to map the O-GlcNAcylation site. b, Quantification of H4 O-GlcNAcylation. WB from Fig. 1a were quantified. n = 4 biologically independent experiments. P values were calculated by unpaired, two-tailed t-tests, means ± s.d., n.s., non-significant. a.u., arbitrary unit. c, Functional assessment of mutations at H4S47 in DNA replication. HEK293T cells were transfected with Flag-H4WT, Flag-H4S47A for 24 h and cultured with or without PUG (100 μM) for another 24 h prior to EdU staining and FACS analysis. d, e, Evaluation of replication efficiency in HT1080 and A549 cells. HT1080 (d) or A549 (e) cells were transfected and treated as in Fig. 1a before the analyse of replication efficiency by immunofluorescence. Representative images (left) and associated quantifications (right) are demonstrated. Cells positive for both PCNA and Flag were marked with dashed lines. From left, n = 1,981, 2,232, 1,553, 1,181 (d) and n = 821, 963, 827, 1,306 (e) cells. P values were calculated by unpaired, two-tailed t-tests. For all box plots, the bottom, middle line and top of the box and whiskers indicate the 25th, 50th, 75th and 10–90th percentiles, respectively and means were shown as red ‘+’ signs. n.s., non-significant. Scale bar, 10 μm. a.u., arbitrary unit.

Source data

Extended Data Fig. 2 H4S47 O-GlcNAcylation influences cell proliferation.

a-c, Impact from H4S47 O-GlcNAcylation on cell proliferation. HEK293T (a), HT1080 (b) and A549 (c) cells were transfected with Flag-H4WT or -H4S47A and incubated with or without PUG (100 μM). Cell numbers were counted every 24 h. n = 3 biologically independent experiments. Two-way ANOVA Tukey’s multiple comparisons test, means ± s.d., n.s., non-significant. d, Functional assessment of mutations at H4S47 in DNA replication. HEK293T cells were transfected with Flag-H4WT, Flag-H4S47A, Flag-H4S47D or Flag-H4S47E for 48 h prior to the detection of replication efficiency by FACS analysis. e, DNA fiber assay for replication fork function. Frequency distribution of CIdU track lengths (left) and inter-origin distances (right) for experiments in Fig. 1d were shown. f, Assessment of DNA damage response. HEK293T cells transfected and treated as in Fig. 1a were analyzed by WB with indicated antibodies. HU (3 mM) treatment for 3 h was included as a positive control.

Source data

Extended Data Fig. 3 H4S47 O-GlcNAcylation does not affect cell viability.

a, b, Analysis of cell survival and apoptosis. HEK293T cells were transfected and treated as in Extended Data Fig. 2a. Etoposide (10 μM) treatment for 24 h was used as a positive control. Following staining with Annexin V and PI, cells were analyzed by flow cytometry. Representative FACS profiles (a) and quantifications of apoptotic cells (b, left) and viable cells (b, right) are demonstrated. n = 3 biologically independent experiments. P values were calculated by Tukey’s multiple comparison test (two-way ANOVA), means ± s.d., n.s., non-significant.

Source data

Extended Data Fig. 4 OGT and OGA distribution during the cell cycle progression.

a, b, Examination of proteins in cells synchronized at different stages of the cell cycle. HEK293T cells synchronized at G1/S were released and collected at indicated time points. Experimental setup and FACS profiles depicting the cell cycle progression were demonstrated (top). Both total proteins (a) and chromatin pellets (b) were analyzed by WB (bottom). Coomassie-stained total histones, H4 and β-actin served as loading controls. Asyn, asynchronization. H4S47ph, H4 phosphorylated at S47.

Source data

Extended Data Fig. 5 OGT promotes DNA replication through H4S47 O-GlcNAcylation.

a, Quantification of H4 O-GlcNAcylation levels. WB from Fig. 2d were quantified. n = 4 biologically independent experiments. P values measured by unpaired, two-tailed t-tests, means ± s.d., n.s., non-significant. a.u., arbitrary unit. b, Evaluation of replication efficiency by EdU incorporation. HEK293T cells were transfected with p3 × Flag-CMV-10 (empty vector) or Flag-OGT for 48 h prior to the detection of replication efficiency with immunofluorescence analysis. HU (3 mM) treatment for 3 h was used as a positive control. Representative images (left) and quantifications of EdU intensities of PCNA-positive cells (right) were demonstrated. From left, n = 1,551, 1,063, 1,234 cells. P values were calculated by unpaired, two-tailed t-tests. For all box plots, the bottom, middle line and top of the box and whiskers indicate the 25th, 50th, 75th and 10–90th percentiles, respectively and means were shown as red ‘+’ signs. Scale bar, 10 μm. a.u., arbitrary unit. c, Analysis of protein O-GlcNAcylation. HEK293T cells were incubated with or without OSMI-1 (50 μM) for 24 h and analyzed by WB with CTD110.6 antibody. d, Examination of EdU intensity by FACS. HEK293T cells were treated as described in (c) and pulsed with EdU for 30 min. HU treatment (3 mM) for 3 h was included as a positive control. e, Representative images from Fig. 2e. Cells positive for Flag (H4) were selected for the quantification of EdU intensities. Scale bar, 10 μm. f, g, DNA fiber assay. Experimental setup (f, top), representative images (f, bottom) as well as frequency distribution of CIdU track lengths (g, left) and inter-origin distances (g, right) for Fig. 2f were shown. Scale bar, 10 μm.

Source data

Extended Data Fig. 6 Functional evaluation of H4S47 O-GlcNAcylation in cells challenged with replication stresses.

a, b, Impact from Glc or Gln deficiency on DNA replication. From representative experiments included in Fig. 2g (a) and Fig. 2h (b), mean intensities of EdU of Flag- and PCNA-positive cells were shown. From left, n = 4,896, 3,249, 4,968, 3,316, 4,488, 3,083, 5,131, 2,413 (a) and n = 8,404, 2,180, 4,397, 2,514, 8,862, 3,090, 7,592, 1,599, 4,413, 1,801 (b) cells. c-f, Influences from HU or CPT on H4 O-GlcNAcylation and DNA replication. HEK293T cells were transfected as in Fig. 1a for 24 h, and then cultured in medium containing HU (c, d) or CPT (e, f) at different concentrations as indicated for another 24 h. Subsequently, cells were subjected to Flag IP for the detection of H4 O-GlcNAcylation (c, e, top) and immunofluorescence as in Fig. 1c for replication efficiency analysis (c, e, bottom). Different samples were loaded on the same gel, but not in adjacent lanes (e, top). Curves (c, e, bottom) based on means of EdU intensities from cells positive for both Flag and PCNA were shown. n = 3 (c, e, bottom) biologically independent experiments. For curves, P values calculated by Sidak’s multiple comparison test (two-way ANOVA) (c, e, bottom). From left, n = 2,074, 2,878, 1,707, 2,467, 1,185, 2,987, 2488, 2,819 (d) and n = 1,714, 2,244, 3,790, 1,214, 1,968, 432, 476, 670 (f) cells. For all box plots, P values were calculated by Tukey’s multiple comparison test (one-way ANOVA of unpaired t-tests); The bottom, middle line and top of the box and whiskers indicate the 25th, 50th, 75th and 10–90th percentiles, respectively and means were shown as ‘+’ signs (a, b, d and f). n.s., non-significant, a.u., arbitrary unit.

Source data

Extended Data Fig. 7 Confirmation of the regulation of MCM complex by H4S47 O-GlcNAcylation.

a, b, Assay for subunits in MCM complex associating with H4. HT1080 (a) or A549 (b) cells were transfected and treated as in Fig. 1a. IgG served as a negative control. Representative experiment (left) and quantification of MCM2/6 levels were shown (right). n = 3 (a), n = 4 (b) biologically independent experiments. P values were calculated by Tukey’s multiple comparison test (two-way ANOVA), means ± s.d., n.s., non-significant. c, d, Examination of MCM2 pS53 in chromatin fraction. HT1080 (c) or A549 (d) cells were transfected and treated as in Fig. 1a. Chromatin pellets were assayed with indicated antibodies. Representative experiment (left) and quantification of MCM2 pS53 levels were shown (right). n = 3 biologically independent experiments. P values were measured by paired, two-tailed t-tests, means ± s.d., n.s., non-significant. a.u., arbitrary unit.

Source data

Extended Data Fig. 8 Confirmation of DDK function in the regulation of DNA replication by H4S47 O-GlcNAcylation.

a, Analysis of CDK2 and Cyclin E association with H4. HEK293T cells were transfected and treated as in Fig. 1a. IgG served as a negative control. b, c, Quantification of DBF4 and CDC7. WB from Fig. 4b (b) and Fig. 4c (c) were quantified, respectively. n = 3 biologically independent experiments. d, In vitro assay of H4 O-GlcNAcylation. His-H4WT and -H4S47A isolated from BL21 competent E. coli, which co-expressed OGT, were analyzed by WB with O-GlcNAcylation antibody CTD110.6. e, Quantification of DBF4, CDC7 and MCM2 pS53 levels for experiments in Fig. 4e. n = 3 biologically independent experiments. f, Overexpression of DBF4 enhances DNA replication. GFP-DBF4 or pEGFP-N1 (empty vector) was transfected along with Flag-H4WT or -H4S47A into HEK293T cells for 48 h prior to the detection of replication efficiency. From left, n = 4,183, 4,882, 3,133, 6,626 cells. P values were calculated by unpaired, two-tailed t-tests. For all box plots, the bottom, middle line and top of the box and whiskers indicate the 25th, 50th, 75th and 10–90th percentiles, respectively and means were shown as red ‘+’ signs. g, Quantification of MCM2/7 and DBF4 for experiments in Fig. 5c. n = 3 biologically independent experiments. h, DNA fiber assay. Experimental setup (left) and frequency distribution of inter-origin distances (right) for Fig. 6a were shown. i, Detection of H4S47 O-GlcNAcylation by mass spectrometry. Tandem mass spectrum of peptides containing S47 O-GlcNAcylation from experiments described in Fig. 6d was shown. The y, a and b fragmentations were used to map the O-GlcNAcylation site. P values calculated by Tukey’s multiple comparison test (two-way ANOVA), means ± s.d., n.s., non-significant (b, c, e and g). a.u., arbitrary unit.

Source data

Extended Data Fig. 9 Genome-wide investigation of H4S47 O-GlcNAcylation function in origin activation.

a, Heat maps of SNS signals. HEK293T cells were prepared as in Fig. 1a. Heat maps demonstrating SNS signals from RNase-treated and untreated samples. n = 37,590 (H4WT), n = 35,625 (H4WT + PUG), n = 29,103 (H4S47A), n = 27,034 (H4WT + PUG) peaks. b, Box plots of SNS signals among H4WT peaks. n = 37,590 peaks. RRPM, reference-adjusted reads per million. P values were calculated by unpaired, two-tailed t-tests using R. For box plots, the bottom, middle line and top of the box and whiskers indicate the 25th, 50th, 75th percentiles and minimum to maximum, respectively. c, Venn diagram showing the overlap among origins of H4WT and origins upregulated by PUG treatment. d, Enriched DNA motifs in replication origins. P values calculated by cumulative binomial distributions. e, Genome-wide distribution of SNS peaks. n = 37,590. UTR, untranslated region. TTS, transcriptional terminate sites. f, Enrichment of MCM6 at replication origins. HEK293T cells incubated with or without PUG (100 μM) for 24 h were assayed by ChIP with antibody against MCM6. The y axis is the ratio of DNA in immunoprecipitate to that in input. n = 4 (TOP1), n = 3 (MCM4), n = 3 (ChrX (ENm006)) biologically independent experiments. g, h, Association of Flag (H4) and MCM6 at replication origins. HEK293T cells transfected and treated as in Fig. 1a were analyzed by ChIP using antibody against Flag (H4) (g) or MCM6 (h). The y axis is the ratio of DNA in the immunoprecipitate to that in the input. n = 4 (g, TOP1), n = 3 (g, MCM4), n = 4 (g, ChrX (ENm006)), n = 3 (h) biologically independent experiments. Two-way ANOVA Tukey’s multiple comparisons test, means ± s.d., n.s., non-significant (f-h).

Source data

Extended Data Fig. 10 Genome-wide evaluation of H4S47 O-GlcNAcylation function in replication timing.

a-i, Functional comparison between H4WT and H4S47A for replication timing. HEK293T and HT1080 cells were transfected as in Fig. 1a (b, d, e, f) and treated with PUG (100 μM) for 24 h (c, g, h, i). Following BrdU (100 μM) pulse-labelling for 2 h, S1, S2, S3 and S4 fractions (a) were isolated for genomic sequencing. b, c, Integrative Genomics Viewer (IGV) snapshots of the Log2 E/L (Early/Late) Repli-seq plots. Raw RT data (Log2 E/L ratio) is shown for a chromosomal region. d, g, Dot plot of replication timing values. RDs (replication timing domains) defined in wild type HEK293T (d, n = 2,493; g, n = 2,579) and HT1080 (d, n = 2,275; g, n = 2,245) were demonstrated. e, f, h, i, Analysis of replication timing values of early/late timing domains. Meta profiles show replication timing values of the length percentage aligned early (e, h) and late (f, i) timing domains. Replication timing domains in HEK293T (e, n = 1,129; h, n = 1,209) and HT1080 (e, n = 1,002; h, n = 963) with replication timing value > 0 are defined as early RDs (e, h, left). Replication timing domains in HEK293T (f, n = 1,356; i, n = 1,389) and HT1080 (f, n = 1,266; i, n = 1,237) with replication timing value < 0 are defined as late RDs (f, i, left). Heatmaps show replication timing values of both early (e, h, right) and late (f, i, right) timing domains in HEK293T and HT1080 cells. Replication timing value was counted 5 Mb upstream and downstream of the centre of each early or late timing domain. RT, replication timing; RD, replication timing domain; Mb, mega base pair; Chr1, chromosome 1.

Supplementary information

Supplementary Information

Supplementary Figures 1–5.

Reporting Summary

Peer Review File

Supplementary Tables 1–4

Key materials, including information on antibodies, chemicals, and reagent kits, primers, and recombinant DNA.

Supplementary Data 1

Statistical source data for Supplementary Figure 1.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Unprocessed western blots and gels.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots and gels.

Source Data Fig. 4

Unprocessed western blots and gels.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots and gels.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots and gels.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical 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

Zou, Y., Pei, J., Long, H. et al. H4S47 O-GlcNAcylation regulates the activation of mammalian replication origins. Nat Struct Mol Biol 30, 800–811 (2023). https://doi.org/10.1038/s41594-023-00998-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-00998-6

This article is cited by

Search

Quick links

Nature Briefing

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

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