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:

Epigenetic priming by Dppa2 and 4 in pluripotency facilitates multi-lineage commitment

Abstract

How the epigenetic landscape is established in development is still being elucidated. Here, we uncover developmental pluripotency associated 2 and 4 (DPPA2/4) as epigenetic priming factors that establish a permissive epigenetic landscape at a subset of developmentally important bivalent promoters characterized by low expression and poised RNA-polymerase. Differentiation assays reveal that Dppa2/4 double knockout mouse embryonic stem cells fail to exit pluripotency and differentiate efficiently. DPPA2/4 bind both H3K4me3-marked and bivalent gene promoters and associate with COMPASS- and Polycomb-bound chromatin. Comparing knockout and inducible knockdown systems, we find that acute depletion of DPPA2/4 results in rapid loss of H3K4me3 from key bivalent genes, while H3K27me3 is initially more stable but lost following extended culture. Consequently, upon DPPA2/4 depletion, these promoters gain DNA methylation and are unable to be activated upon differentiation. Our findings uncover a novel epigenetic priming mechanism at developmental promoters, poising them for future lineage-specific activation.

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: DPPA2/4 are required for differentiation and bind developmental bivalent promoters.
Fig. 2: DPPA2/4 are required to maintain bivalent chromatin at a subset of developmental genes.
Fig. 3: DPPA2/4-dependent bivalent genes characterized by low H3K4me3, low expression and initiating but not elongating RNA polymerase II.
Fig. 4: DPPA2/4-dependent promoters gain DNA methylation and fail to be upregulated during differentiation.
Fig. 5: DPPA2/4 are required to target H3K4me3 and prevent DNA methylation at a subset of bivalent genes.
Fig. 6: DPPA2/4 prime the epigenetic landscape at bivalent genes, facilitating cell differentiation.

Similar content being viewed by others

Data availability

All sequencing data generated in this study have been submitted to the Gene Expression Omnibus under accession number GSE135841. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE38 partner repository with the data set identifier PXD014981. DPPA2 and DPPA4 ChIP data were reanalyzed from ref. 9 (GSE117173), RNA polymerase II ChIP was reanalyzed from ref. 20 (GSE34520), EZH2 and SUZ12 ChIP data in Fig. 2 were reanalyzed from ref. 39 (GSE49435), ASH2L-GFP ChIP data in Fig. 2 reanalyzed from ref. 1 (GSE52071), MLL2 ChIP data in Fig. 2 reanalyzed from ref. 2 (GSE48172), TIP60/KAT5 ChIP data reanalyzed from ref. 40 (GSE69671), high-confidence bivalent gene list from ref. 14, DNMT TKO ESC RNA-seq data from ref. 21 (GSE67867) and 2C-like ZGA gene list from ref. 41. Uncropped images of immunoblots presented in Fig. 5b are provided as source data.

References

  1. Denissov, S. et al. Mll2 is required for H3K4 trimethylation on bivalent promoters in embryonic stem cells, whereas Mll1 is redundant. Dev. 141, 526–537 (2014).

    Article  CAS  Google Scholar 

  2. Hu, D. et al. The Mll2 branch of the COMPASS family regulates bivalent promoters in mouse embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1093–1097 (2013).

    Article  CAS  Google Scholar 

  3. Voigt, P., Tee, W.-W. & Reinberg, D. A double take on bivalent promoters. Genes Dev. 27, 1318–1338 (2013).

    Article  CAS  Google Scholar 

  4. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  Google Scholar 

  5. Azuara, V. et al. Chromatin signatures of pluripotent cell lines. Nat. Cell Biol. 8, 532–538 (2006).

    Article  CAS  Google Scholar 

  6. Eckersley-Maslin, M. et al. Dppa2 and Dppa4 directly regulate the Dux-driven zygotic transcriptional program. Genes Dev. 33, 194–208 (2019).

    Article  CAS  Google Scholar 

  7. De Iaco, A., Coudray, A., Duc, J. & Trono, D. DPPA2 and DPPA4 are necessary to establish a 2C‐like state in mouse embryonic stem cells. EMBO Rep. 20, e47382 (2019).

  8. Yan, Y.-L. et al. DPPA2/4 and SUMO E3 ligase PIAS4 opposingly regulate zygotic transcriptional program. PLoS Biol. 17, e3000324 (2019).

    Article  Google Scholar 

  9. Hernandez, C. et al. Dppa2/4 facilitate epigenetic remodeling during reprogramming to pluripotency. Cell. Stem Cell 23, 396–411.e8 (2018).

    Article  CAS  Google Scholar 

  10. Klein, R. H., Tung, P.-Y., Somanath, P., Fehling, H. J. & Knoepfler, P. S. Genomic functions of developmental pluripotency associated factor 4 (Dppa4) in pluripotent stem cells and cancer. Stem Cell Res. 31, 83–94 (2018).

    Article  CAS  Google Scholar 

  11. Engelen, E. et al. Proteins that bind regulatory regions identified by histone modification chromatin immunoprecipitations and mass spectrometry. Nat. Commun. 6, 7155 (2015).

    Article  Google Scholar 

  12. Madan, B. et al. The pluripotency-associated gene Dppa4 is dispensable for embryonic stem cell identity and germ cell development but essential for embryogenesis. Mol. Cell. Biol. 29, 3186–3203 (2009).

    Article  CAS  Google Scholar 

  13. Nakamura, T., Nakagawa, M., Ichisaka, T., Shiota, A. & Yamanaka, S. Essential roles of ECAT15-2/Dppa2 in functional lung development. Mol. Cell. Biol. 31, 4366–4378 (2011).

    Article  CAS  Google Scholar 

  14. Mas, G. et al. Promoter bivalency favors an open chromatin architecture in embryonic stem cells. Nat. Genet. 50, 1452–1462 (2018).

    Article  CAS  Google Scholar 

  15. Maldonado-Saldivia, J. et al. Dppa2 and Dppa4 are closely linked SAP motif genes restricted to pluripotent cells and the germ line. Stem Cells 25, 19–28 (2007).

    Article  CAS  Google Scholar 

  16. Masaki, H., Nishida, T., Sakasai, R. & Teraoka, H. DPPA4 modulates chromatin structure via association with DNA and core histone H3 in mouse embryonic stem cells. Genes Cells 15, 327–337 (2010).

    Article  CAS  Google Scholar 

  17. Papachristou, E. K. et al. A quantitative mass spectrometry-based approach to monitor the dynamics of endogenous chromatin-associated protein complexes. Nat. Commun. 9, 2311 (2018).

    Article  Google Scholar 

  18. Surface, L. E. et al. H2A.Z.1 monoubiquitylation antagonizes BRD2 to maintain poised chromatin in ESCs. Cell Rep. 14, 1142–1155 (2016).

    Article  CAS  Google Scholar 

  19. Benayoun, B. A. et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 158, 673–688 (2014).

    Article  CAS  Google Scholar 

  20. Brookes, E. et al. Polycomb associates genome-wide with a specific RNA polymerase II variant, and regulates metabolic genes in ESCs. Cell Stem Cell 10, 157–170 (2012).

    Article  CAS  Google Scholar 

  21. Domcke, S. et al. Competition between DNA methylation and transcription factors determines binding of NRF1. Nature 528, 575–579 (2015).

    Article  CAS  Google Scholar 

  22. Klobucar, T. et al. IMPLICON: an ultra-deep sequencing method to uncover DNA methylation at imprinted regions. Preprint at bioRxiv https://doi.org/10.1101/2020.03.21.000042 (2020).

  23. Sneppen, K. & Ringrose, L. Theoretical analysis of Polycomb-Trithorax systems predicts that poised chromatin is bistable and not bivalent. Nat. Commun. 10, 2133 (2019).

    Article  Google Scholar 

  24. Ooi, S. K. T. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  Google Scholar 

  25. Creyghton, M. P. et al. The histone variant H2AZ is enriched at Polycomb group target genes in ES cells and is necessary for proper execution of developmental programs. Cell 135, 649 (2008).

    Article  CAS  Google Scholar 

  26. Ku, M. et al. H2A.Z landscapes and dual modifications in pluripotent and multipotent stem cells underlie complex genome regulatory functions. Genome Biol. 13, R85 (2012).

    Article  CAS  Google Scholar 

  27. Wang, Y. et al. Histone variants H2A.Z and H3.3 coordinately regulate PRC2-dependent H3K27me3 deposition and gene expression regulation in mES cells. BMC Biol. 16, 107 (2018).

    Article  Google Scholar 

  28. Hu, G. et al. H2A.Z facilitates access of active and repressive complexes to chromatin in embryonic stem cell self-renewal and differentiation. Cell Stem Cell 12, 180–192 (2013).

    Article  CAS  Google Scholar 

  29. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  30. Ficz, G. et al. FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13, 351–359 (2013).

    Article  CAS  Google Scholar 

  31. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  Google Scholar 

  32. Kirschner, K. et al. Phenotype specific analyses reveal distinct regulatory mechanism for chronically activated p53. PLoS Genet. 11, e1005053 (2015).

    Article  Google Scholar 

  33. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  Google Scholar 

  34. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

  35. Pintacuda, G. et al. hnRNPK recruits PCGF3/5-PRC1 to the Xist RNA B-repeat to establish Polycomb-mediated chromosomal silencing. Mol. Cell 68, 955–969.e10 (2017).

    Article  CAS  Google Scholar 

  36. Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).

    Article  CAS  Google Scholar 

  37. Witten, I. H. et al. Data Mining: Practical Machine Learning Tools And Techniques (Morgan Kaufmann, 2016).

  38. Vizcaíno, J. A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, 11033–11033 (2016).

    Article  Google Scholar 

  39. Kaneko, S., Son, J., Shen, S. S., Reinberg, D. & Bonasio, R. PRC2 binds active promoters and contacts nascent RNAs in embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1258–1264 (2013).

    Article  CAS  Google Scholar 

  40. Ravens, S., Yu, C., Ye, T., Stierle, M. & Tora, L. Tip60 complex binds to active Pol II promoters and a subset of enhancers and co-regulates the c-Myc network in mouse embryonic stem cells. Epigenet. Chromatin 8, 45 (2015).

    Article  Google Scholar 

  41. Eckersley-Maslin, M. A. et al. MERVL/Zscan4 network activation results in transient genome-wide DNA demethylation of mESCs. Cell Rep. 17, 179–192 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Reik laboratory for helpful discussions. We also thank F. di Tullio for help generating overexpression cell lines, F. Krueger for processing sequencing data and for general bioformatics support, S. Wingett for bioinformatic assistance and S. Andrews for bioinformatic advice. We thank B. Hussey and E. Easthope at Sanger Institute and K. Tabbada at Babraham Institute for assistance with high-throughput sequencing, R. Walker for assistance with flow cytometry, and J. Webster and D. Oxley for mass spectrometry. Dnmt TKO cells were a kind gift from D. Schübeler (FMI). M.A.E.-M. is supported by a BBSRC Discovery Fellowship (BB/T009713/1) and was supported by an EMBO Fellowship (ALTF938–2014) and a Marie Sklodowska-Curie Individual Fellowship. A.P. is supported by a Sir Henry Wellcome Fellowship (215912/Z/19/Z). M.B. was supported by an Erasmus Grant. M.N. and Y.I. were supported by a Cancer Research UK Cambridge Institute Core grant (no. C9545/A29580). Research in the Reik laboratory is supported by the Biotechnology and Biological Sciences Research Council (BB/K010867/1) and the Wellcome Trust (095645/Z/11/Z).

Author information

Authors and Affiliations

Authors

Contributions

M.A.E.-M. and W.R. conceived, designed and supervised the study. M.A.E.-M. performed experiments, analyzed data and wrote the paper. A.P. helped perform ChIP–qPCR, ChIP–seq, CoIP, inducible knockdown and western blotting experiments, prepared cells for proteomics and analyzed proteomic and ChIP–seq data. M.B. performed embryoid body differentiation assays and qPCRs. C.K. analyzed chomatin state association of DPPA2/4 and generated the random forest model for DPPA2/4 dependency based on promoter features. V.N.R.F. and C.S.D’S. performed qPLEX-RIME experiments. Y.I. and M.N. designed inducible shRNA vectors.

Corresponding authors

Correspondence to Mélanie A. Eckersley-Maslin or Wolf Reik.

Ethics declarations

Competing interests

W.R. is a consultant and shareholder of Cambridge Epigenetix. The remaining authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available. Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 DPPA2/4 are required for differentiation related to Fig. 1.

a, Western Blotting showing knockout of DPPA2 (top) and DPPA4 (middle) protein in WT (left 3 columns) and Dppa2/4 double knockout (DKO, right 3 columns) cells. β-ACTIN is shown as loading control b, Expression (Log-2 reads per million) of core (orange), naïve (blue), formative (purple) and primed (aqua) pluripotency markers in WT (black) and Dppa2/4 DKO (blue) ESCs as determined by RNA-sequencing. Plotted is the mean + /- standard deviation for 3 individual WT or DKO clones, each prepared in technical triplicate except for DKO clone 43 which was prepared in duplicate (n = 9 WT and 8 DKO total). An individual unpaired two-tailed t-test was performed for each gene to give the indicated p-values (above each pair of bars) where significant (p < 0.05) values are bold. c, Representative immunofluorescence staining of wild type (WT, 3 individual clones) and Dppa2/4 double knockout (DKO, three individual clones) cells showing OCT4 (top), NANOG (middle, red) and SOX2 (bottom, cyan) pluripotency markers. DAPI is included as a DNA counterstain. Scale bar represents 50μm. dg, Quantitative RT-PCR of pluripotency d, ectoderm e, endoderm f, and mesoderm g, markers during 9-day embryoid body differentiation between wild type (WT, gray, clone 58) and Dppa2/4 DKO (DKO, colored, clone 43) cells. Error bars represent mean plus standard deviation of three independent differentiation experiments. RNA samples are the same that were used for RNA sequencing in Fig. 1a, b.

Extended Data Fig. 2 DPPA2/4 bind developmental bivalent promoters, related to Fig. 1.

a, Genome browser screenshot showing DPPA2 (blue), DPPA4 (purple) binding and H3K4me3 (green) and H3K27me3 (red) histone modifications across a 394 kb region of chromosome 1. Input, DPPA2 and DPPA4 data reanalyzed from9 [GSE117173], H3K4me3 and H3K27me3 data from this study. H3K4me3 promoters highlighted in green, bivalent promoters highlighted in yellow, inactive promoters highlighted in red. b, Distribution of high confidence DPPA2 and 4 peaks. Data reanalyzed from9 [GSE117173]. c, Log2 enrichment of DPPA2 (blue) and DPPA4 (purple) binding at gene promoters for different expression bins. H3K4me3 enrichment (green) is shown for comparison. d, Western blot showing levels of overexpression of DPPA2-GFP (top) and DPPA4-GFP (middle) in different stable clonal cell lines. β-ACTIN (bottom) is shown as loading control. Open arrows denote endogenous protein, filled arrows overexpressed GFP-tagged protein. e,f, Scatter plots showing gene expression between ESCs stably overexpressing GFP and DPPA2-GFP e, or DPPA4-GFP f, (two independent cellular clones each harvested in triplicate, separated by at least one passage). Differentially expressed genes (calculated using DESeq p-value <0.05 after Benjamimi and Hochberg correction) are shown in blue, differentially expressed 2C-like ZGA genes are shown in red. Dppa2 and Dppa4 are shown in green. g, Expression of core, naïve, formative and primed pluripotency markers in E14 + GFP (black, grey), E14 + DPPA2-GFP (blue) and E14-DPPA4-GFP (purple) overexpression clones as determined by RNA-sequencing. Two independent cellular clones per overexpression or control line were harvested in triplicate, separated by at least one passage. h, Overlap in differentially expressed genes in DPPA2-GFP (blue) and DPPA4-GFP (purple) stable overexpression clones with 2C-like (left, orange) and bivalent (right, green) genes. i, Expression of 2C-like genes, bivalent genes and proteomics hits in GFP (grey), DPPA2-GFP (blue), DPPA4-GFP (purple) stable overexpressing cells. 2C-like genes from 48, bivalent genes from13. Two independently targeted clones (containing the average of n = 3 replicates harvested independently) are shown per condition in all subpanels. The central line denotes the median, the yellow box the 25th and 75th percentile of the data and the black whiskers the median + /- the interquartile range (25–75%) multiplied by 2. Circles represent single promoters that fall outside this range. j, qPLEX-RIME results showing log2fold change between DPPA2-GFP and GFP control versus log10 (adjusted p-value). Dotted line represents cut off of p < 0.05. DPPA2 and DPPA4 are shown in orange. Top 3 interactors are shown in black. Members of Polycomb (red), COMPASS (green) and SRCAP/INO-80 (blue) complexes are highlighted. k, Western Blotting showing levels of H3K4me3 (top) and H3K27me3 (middle) in different stable cell lines. β-ACTIN is shown as a loading control. l, Endogenous DPPA4 immunoprecipitation (IP) followed by Western Blot in WT (first two columns) and Dppa2/4 DKO (last two columns) cells. * denotes IgG band. m, Aligned probe plots of a random selection of 5000 genes showing enrichment of different chromatin proteins ordered by DPPA4 levels. Input, DPPA2 and DPPA4 ChIP data reanalysed from9 [GSE11717], ASH2L-GFP data from 1 [GSE52071], MLL2 data from2 [GSE48172], EZH2 and SUZ12 data from39 [GSE49435] and KAT5/TIP60 data from30 [GSE69671].

Extended Data Fig. 3 DPPA2/4 are required to maintain bivalent chromatin at a subset of developmental genes, related to Fig. 2.

a, Western blot showing protein levels of chromatin factors and histone modifications between three wild type (WT) ESC clones (left) and three Dppa2/4 DKO ESC cellular clones (right). Histone H3 and β-ACTIN are included as loading controls. b, Scatter plots showing enrichment at gene promoters for H3K4me3, H3K27me3, H2A.Z, RING1B, EZH2 and ASH2L between WT (x-axis) and Dppa2/4 DKO (y-axis) ESCs. Protein enrichment at DPPA2/4-dependent (apricot), -sensitive (green) and -dependent (teal) genes are shown. c, Box-whisker plots showing enrichment of different chromatin proteins, histone modifications and chromatin accessibility measured by ATAC-seq at DPPA2/4-dependent (left, apricot), -sensitive (middle, green) and -independent (right, blue) promoters (TSS + /-1kb) comparing WT (light) and Dppa2/4 DKO (dark) ESCs. Input, DPPA2 and DPPA4 ChIP data from8. All other data from this study (n = 3 cellular clones for H3K4me3 and H3K27me3 (pooled data) and 1 cellular clone for other marks). The central line denotes the median, the yellow box the 25th and 75th percentile of the data and the black whiskers the median + /- the interquartile range (25–75%) multiplied by 2. Circles represent single promoters that fall outside this range. d, ChIP-qPCR results at DPPA2/4-dependent genes Twist2 and Fermt1, compared to control Klf4 gene promoters in 2–3 replicates (see data points, replicates harvested on different days) of one cell clone each of WT (grey) and Dppa2, 4 DKO (blue) cells for H3K4me3, H3K27me3, H2A.Z, EZH2, RING1B and IgG control. Error bars represent mean + standard deviation. Individual data points are shown. Unpaired two-sided t-tests were used and p-values are shown. e, f, Genome browser view of a DPPA2/4-dependent gene (Fermt1) e, and a -dependent (Nphs2) and -independent (Tdrd5) gene f, showing enrichment of chromatin modifications, proteins, chromatin accessibility (blue), DNA methylation (green/grey) and transcription (bottom two panels) for wild type (WT) and Dppa2/4 double knockout (DKO) ESCs. g, Accessibility measured by ATAC-seq at gene promoters (TSS + /-1kb) comparing DPPA2/4-dependent (apricot), -sensitive (green) and -independent (teal) promoters between WT (x-axis) and Dppa2/4 DKO (y-axis) cells.

Extended Data Fig. 4 DPPA2/4-dependent bivalent genes characterized by low H3K4me3, low expression and initiating but not elongating RNA polymerase II, related to Fig. 3.

a, Scatterplot comparing H3K4me3 peak width (x-axis) and expression of associated gene (y-axis) for Dppa2/4-dependent (orange), -sensitive (green) and -independent (blue) genes (as defined in Fig. 2c). H3K4me3 data is from 3 independent cellular clones. b, Enrichment for different RNA polymerase II modifications across gene bodies. Data reanalysed from20 [GSE34520]. Line indicates the median. c, Enrichment of different chromatin modifications and chromatin proteins along with chromatin accessibility (ATAC-seq) between Dppa2/4-dependent (orange), -sensitive (green) and -independent (blue) gene promoters (TSS + /-1kb). DPPA2 and 4 ChIP-seq data reanalysed from9 [GSE117173], all other data from this study. Line indicates the median value. d, proportion of gene promoters (start of gene + /- 1 kb) sequence masked by repeats. Line indicates the median value. e, CpG density, GC content and DNA methylation levels at Dppa2/4-dependent (orange), -sensitive (green) and -independent (blue) gene promoters (TSS + /-1kb). Line indicates the median value.

Extended Data Fig. 5 DPPA2/4-dependent promoters gain DNA methylation and fail to be upregulated during differentiation, related to Fig. 4.

a, Box-whisker plots of the lowest 20% of expressed genes in WT ESCs (total number 3460), showing all 3460 genes, excluding DPPA2/4-dependent and -sensitive genes, only DPPA2/4-dependent genes and only DPPA2/4 sensitive genes during the embryoid body time course between WT (grey) and DKO (blue) cells. Day of differentiation is denoted below each pair of bars. The central line denotes the median, the yellow box the 25th and 75th percentile of the data and the black whiskers the median + /- the interquartile range (25–75%) multiplied by 2. Circles represent single promoters that fall outside this range. Each bar shows pooled data from three differentiation timecourses ran on different days using 1 WT and 1 DKO clone. b, Mass spectrometry quantification of 5-methylcytosine (5mC, left) and 5-hydroxymethylcytosine (5hmC, right) between WT (grey) and Dppa2/4 DKO (blue) cells. Bars represent mean + /- standard deviation of three independent cellular clones. c, Amplicon-bisulfite-seq analysis of DPPA2/4-dependent gene promotors and 2 control regions (Klf4 and Sox2) in three wild type (WT, first three columns) and Dppa2/4 DKO (last 3 columns) clones. Each row represents an individual CpG included within the amplicon. White rows represent break between different amplicon regions (between 1 and 3 per gene promoter analysed). A minimum of 100-fold coverage per CpG was required to include the CpG in the analysis. d, Mass spectrometry quantification of 5-methylcytosine (5mC, left) and 5-hydroxymethylcytosine (5hmC, right) between Dnmt WT (left two columns) and Dnmt TKO (right two columns) cells following control (grey) or Dppa2/4 siRNA (red) treatment for 4 days. Bars represent mean + /- standard deviation of two replicate experiments harvested on different days. e, Western blot showing levels of DNA methylation machinery (DNMT1, DNMT3A and DNMT3B), DPPA2 and DPPA4 between Dnmt WT (left two columns) and Dnmt TKO (right two columns) following treatment with control siRNA or both Dppa2 and Dppa4 siRNA for 4 days. HSP90 is shown as loading control. f, Expression levels of DPPA2/4-dependent (apricot), -sensitive (green) and -independent (blue) genes as defined in Fig. 2c between Dnmt WT and Dnmt TKO cells. Data reanalysed from21 [GSE67867]. Box plots shown are as described in A. (G) Expression of Dppa2 and Dppa4 mRNAs following control (grey) or Dppa2/4 (red) siRNA treatment in WT (left, dark) and Dnmt TKO (right, light) cells. Relative expression is normalised to the level of control siRNA (dark bars) for WT and Dnmt TKO cells. Dots represent replicates experiments harvested on different days and bars averages plus standard deviation.

Extended Data Fig. 6 DPPA2/4 are required to target H3K4me3 and prevent DNA methylation at a subset of bivalent genes, related to Fig. 5.

a, Western blot for H3K4me3 (top), H3K27me3 (middle) and β-ACTIN (bottom) loading control in inducible shRNA experiment for untreated (-dox, first and fourth column), dox treated (+dox, second and fifth column) and recovery (rec, third and sixth column) cells containing inducible shRNA against Dppa2 (left three columns) or Dppa4 (right three columns). H3K4me3 and H3K27me3 bands are quantified relative to β-ACTIN (below). b, Scatterplots of H3K4me3 peaks (top row) and H3K27me3 peaks (bottom row) for cells containing shRNA against Dppa2 (left two columns) or Dppa4 (right column) highlighting those peaks that are significantly up (blue) or down (red) regulated in +dox versus -dox conditions for the given hairpin. Data is from three replicate experiments harvested on different days using one bulk cell population per inducible hairpin. c, overlap of downregulated H3K4me3 (left) and H3K27me3 (right) peaks between shRNA for Dppa2 (orange) and Dppa4 (green). d, genomic features associated with H3K4me3 (left) and H3K27me3 (right) peaks significantly downregulated with both Dppa2 and Dppa4 shRNAs. e, Table showing number of differentially enriched promoters (DESeq2) and number up- and down-regulated promoters in inducible shRNA experiments. f, Amplicon-bisulfite-seq analysis of DPPA2/4-dependent gene promotors and 2 control regions (Klf4 and Sox2) in inducible shRNA experiment for untreated (-dox, first four columns), dox treated (+dox, middle four columns) and recovery (rec, last four columns) cells containing inducible shRNA against Dppa2 (2) or Dppa4 (4). Two replicate experiments harvested on different days are included per condition. Each row represents an individual CpG included within the amplicon. Crossed rows represent break between different amplicon regions (between 1 and 3 per gene promoter analysed) and crossed columns when insufficient coverage was obtained to accurately measure DNA methylation levels. A minimum of 100-fold coverage per CpG was required to include the CpG in the analysis.

Supplementary information

Reporting Summary

Peer Review Information

Supplementary Table 1

List of all genes, chromosome coordinates, strand and average log2 RPM for embryoid body (EB) differentiation experiments. Three independent differentiations were performed (EBset1, EBset2, EBset3) for one wildtype (clone 58) and one Dppa2/4 DKO (clone 43) clone, and samples were collected at days 1–4, 7 and 9 of differentiation.

Supplementary Table 2

List of differentially expressed genes (DESeq2 + intensity difference filter) for DPPA2-GFP and DPPA4-GFP overexpression lines compared to GFP controls. Columns contain gene name, chromosome coordinates, strand information and classification as differentially expressed in DPPA2 overexpressed cells (Dppa2 O/E, blue), DPPA4-GFP overexpressed cells (Dppa4 O/E, purple), or both DPPA2 and DPPA4 overexpressed cells (Dppa2 O/E and Dppa4 O/E). log2 RPM are give for three biological replicates (_1, _2, _3) for two biological clones for GFP (E14 + GFP_c1, E14 + GFP_c2), DPPA2-GFP (E14 + Dppa2_c3, E14 + Dppa2_c5) or DPPA4-GFP (E14 + Dppa4_c5, E14 + Dppa4_c6) overexpressing cells.

Supplementary Table 3

qPLEX-RIME results showing accession, gene name, description, gene symbol, number of unique peptides detected, peptide intensity for each sample (two biological clones per condition with 3–4 technical replicates each), log2 fold change, average intensity, t, P value, adjusted P value and B value for DPPA-GFP versus GFP (first tab) versus DPPA-GFP versus GFP (second tab).

Supplementary Table 4

List of DPPA2/4-dependent (first tab), DPPA2/4-sensitive (second tab) and DPPA2/4-independent (third tab) gene promoters (TSS ± 1 kb) showing chromosome coordinates, and log2 RPM enrichment for Input, DPPA2, DPPA4, H3K4me3, H3K27me3, H2A.Z, RING1B, ASH2L, EZH2 and chromosome accessibility for WT and DKO cells. Individual clonal replicates are shown. Input, DPPA2 and DPPA4 data reanalyzed from ref. 9, all other data from this study.

Supplementary Table 5

List of attributes used in machine learning, together with their description and data source (first tab) and values (second tab).

Supplementary Table 6

List of differentially enriched H3K4me3 (first tab) and H3K27me3 (second tab) peaks in inducible shRNA experiments for either Dppa2 or Dppa4, showing chromosome coordinates, closest gene within 2 kb, distance to closest gene and log2 RPM values for H3K27me3 and H3K4me3 for all replicates for all conditions.

Supplementary Table 7

Amplicon-bisulfite-sequencing results listing mm10 genomic coordinates of CpG analyzed (columns A–D), gene promoter targeted (column E) average DNA methylation value for inducible shRNA samples (columns G–R), wild-type ESC clones (columns S–U) and Dppa2/4 double knockout cells (columns V–X), and read count for inducible shRNA samples (columns Z–AK), wild-type ESC clones (columns AL–AN) and Dppa2/4 double knockout cells (columns AO–AQ). Each row represents an individual CpG. In red text are CpGs that are omitted due to poor coverage (<100 reads) or quality as they are at the ends of the amplicons. Amplicons are separated by thick borders.

Source data

Source Data Fig. 5

Unprocessed western blots associated with Fig. 5b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Eckersley-Maslin, M.A., Parry, A., Blotenburg, M. et al. Epigenetic priming by Dppa2 and 4 in pluripotency facilitates multi-lineage commitment. Nat Struct Mol Biol 27, 696–705 (2020). https://doi.org/10.1038/s41594-020-0443-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-020-0443-3

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