Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Epigenetic reprogramming enables the transition from primordial germ cell to gonocyte

This article has been updated

Abstract

Gametes are highly specialized cells that can give rise to the next generation through their ability to generate a totipotent zygote. In mice, germ cells are first specified in the developing embryo around embryonic day (E) 6.25 as primordial germ cells (PGCs)1. Following subsequent migration into the developing gonad, PGCs undergo a wave of extensive epigenetic reprogramming around E10.5–E11.52,3,4,5,6,7,8,9,10,11, including genome-wide loss of 5-methylcytosine2,3,4,5,7,8,9,10,11. The underlying molecular mechanisms of this process have remained unclear, leading to our inability to recapitulate this step of germline development in vitro12,13,14. Here we show, using an integrative approach, that this complex reprogramming process involves coordinated interplay among promoter sequence characteristics, DNA (de)methylation, the polycomb (PRC1) complex and both DNA demethylation-dependent and -independent functions of TET1 to enable the activation of a critical set of germline reprogramming-responsive genes involved in gamete generation and meiosis. Our results also reveal an unexpected role for TET1 in maintaining but not driving DNA demethylation in gonadal PGCs. Collectively, our work uncovers a fundamental biological role for gonadal germline reprogramming and identifies the epigenetic principles of the PGC-to-gonocyte transition that will help to guide attempts to recapitulate complete gametogenesis in vitro.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: 5mC and 5hmC dynamics during epigenetic reprogramming.
Figure 2: TET1 safeguards but does not drive DNA demethylation.
Figure 3: GRR genes.
Figure 4: Epigenetic principles of GRR gene activation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 14 March 2018

    The received date was corrected in the HTML from 21 July 2015 to 21 July 2017.

References

  1. Lesch, B. J. & Page, D. C. Genetics of germ cell development. Nat. Rev. Genet. 13, 781–794 (2012)

    Article  CAS  Google Scholar 

  2. Guibert, S., Forné, T. & Weber, M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 22, 633–641 (2012)

    Article  CAS  Google Scholar 

  3. Hackett, J. A. et al. Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine. Science 339, 448–452 (2013)

    Article  ADS  CAS  Google Scholar 

  4. Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008)

    Article  ADS  CAS  Google Scholar 

  5. Hajkova, P. et al. Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15–23 (2002)

    Article  CAS  Google Scholar 

  6. Hill, P. W., Amouroux, R. & Hajkova, P. DNA demethylation, Tet proteins and 5-hydroxymethylcytosine in epigenetic reprogramming: an emerging complex story. Genomics 104, 324–333 (2014)

    Article  CAS  Google Scholar 

  7. Lee, J. et al. Erasing genomic imprinting memory in mouse clone embryos produced from day 11.5 primordial germ cells. Development 129, 1807–1817 (2002)

    Article  CAS  Google Scholar 

  8. Seisenberger, S. et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol. Cell 48, 849–862 (2012)

    Article  CAS  Google Scholar 

  9. Yamaguchi, S. et al. Tet1 controls meiosis by regulating meiotic gene expression. Nature 492, 443–447 (2012)

    Article  ADS  CAS  Google Scholar 

  10. Yamaguchi, S., Shen, L., Liu, Y., Sendler, D. & Zhang, Y. Role of Tet1 in erasure of genomic imprinting. Nature 504, 460–464 (2013)

    Article  ADS  CAS  Google Scholar 

  11. Hajkova, P. et al. Genome-wide reprogramming in the mouse germ line entails the base excision repair pathway. Science 329, 78–82 (2010)

    Article  ADS  CAS  Google Scholar 

  12. Hayashi, K. et al. Offspring from oocytes derived from in vitro primordial germ cell-like cells in mice. Science 338, 971–975 (2012)

    Article  ADS  CAS  Google Scholar 

  13. Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S. & Saitou, M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519–532 (2011)

    Article  CAS  Google Scholar 

  14. Hikabe, O. et al. Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature 539, 299–303 (2016)

    Article  ADS  Google Scholar 

  15. Sun, Z. et al. High-resolution enzymatic mapping of genomic 5-hydroxymethylcytosine in mouse embryonic stem cells. Cell Reports 3, 567–576 (2013)

    Article  CAS  Google Scholar 

  16. Huang, Y. et al. The behaviour of 5-hydroxymethylcytosine in bisulfite sequencing. PLoS One 5, e8888 (2010)

    Article  ADS  Google Scholar 

  17. Yamaguchi, S. et al. Dynamics of 5-methylcytosine and 5-hydroxymethylcytosine during germ cell reprogramming. Cell Res. 23, 329–339 (2013)

    Article  CAS  Google Scholar 

  18. Dawlaty, M. M. et al. Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell 9, 166–175 (2011)

    Article  CAS  Google Scholar 

  19. Weber, M. et al. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat. Genet. 39, 457–466 (2007)

    Article  CAS  Google Scholar 

  20. Tahiliani, M. et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930–935 (2009)

    Article  ADS  CAS  Google Scholar 

  21. Williams, K. et al. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 473, 343–348 (2011)

    Article  ADS  CAS  Google Scholar 

  22. Vella, P. et al. Tet proteins connect the O-linked N-acetylglucosamine transferase Ogt to chromatin in embryonic stem cells. Mol. Cell 49, 645–656 (2013)

    Article  CAS  Google Scholar 

  23. Tsumura, A. et al. Maintenance of self-renewal ability of mouse embryonic stem cells in the absence of DNA methyltransferases Dnmt1, Dnmt3a and Dnmt3b. Genes Cells 11, 805–814 (2006)

    Article  CAS  Google Scholar 

  24. Hargan-Calvopina, J. et al. Stage-specific demethylation in primordial germ cells safeguards against precocious differentiation. Dev. Cell 39, 75–86 (2016)

    Article  CAS  Google Scholar 

  25. Mansour, A. A. et al. The H3K27 demethylase Utx regulates somatic and germ cell epigenetic reprogramming. Nature 488, 409–413 (2012)

    Article  ADS  CAS  Google Scholar 

  26. Yokobayashi, S. et al. PRC1 coordinates timing of sexual differentiation of female primordial germ cells. Nature 495, 236–240 (2013)

    Article  ADS  CAS  Google Scholar 

  27. Ismail, I. H., McDonald, D., Strickfaden, H., Xu, Z. & Hendzel, M. J. A small molecule inhibitor of polycomb repressive complex 1 inhibits ubiquitin signaling at DNA double-strand breaks. J. Biol. Chem. 288, 26944–26954 (2013)

    Article  CAS  Google Scholar 

  28. Deplus, R. et al. TET2 and TET3 regulate GlcNAcylation and H3K4 methylation through OGT and SET1/COMPASS. EMBO J. 32, 645–655 (2013)

    Article  CAS  Google Scholar 

  29. Amouroux, R. et al. De novo DNA methylation drives 5hmC accumulation in mouse zygotes. Nat. Cell Biol. 18, 225–233 (2016)

    Article  CAS  Google Scholar 

  30. Hon, G. C. et al. 5mC oxidation by Tet2 modulates enhancer activity and timing of transcriptome reprogramming during differentiation. Mol. Cell 56, 286–297 (2014)

    Article  CAS  Google Scholar 

  31. Yang, H. et al. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 154, 1370–1379 (2013)

    Article  CAS  Google Scholar 

  32. Boyle, P. et al. Gel-free multiplexed reduced representation bisulfite sequencing for large-scale DNA methylation profiling. Genome Biol. 13, R92 (2012)

    Article  CAS  Google Scholar 

  33. Kurimoto, K. et al. Quantitative dynamics of chromatin remodeling during germ cell specification from mouse embryonic stem cells. Cell Stem Cell 16, 517–532 (2015)

    Article  CAS  Google Scholar 

  34. Borgel, J. et al. Targets and dynamics of promoter DNA methylation during early mouse development. Nat. Genet. 42, 1093–1100 (2010)

    Article  CAS  Google Scholar 

  35. Yu, M. et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell 149, 1368–1380 (2012)

    Article  CAS  Google Scholar 

  36. Xu, Y. et al. Genome-wide regulation of 5hmC, 5mC, and gene expression by Tet1 hydroxylase in mouse embryonic stem cells. Mol. Cell 42, 451–464 (2011)

    Article  CAS  Google Scholar 

  37. 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 

  38. Cooper, S. et al. Targeting polycomb to pericentric heterochromatin in embryonic stem cells reveals a role for H2AK119u1 in PRC2 recruitment. Cell Reports 7, 1456–1470 (2014)

    Article  CAS  Google Scholar 

  39. Hashimoto, H. et al. Structure of a Naegleria Tet-like dioxygenase in complex with 5-methylcytosine DNA. Nature 506, 391–395 (2014)

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Elliot and T. Adejumo for help with fluorescence activated cell sorting, L. Game for help with next-generation sequencing, F. Krueger for providing consensus repetitive element sequences, M. Woodberry, A. Cameron, and J. Glegola for mouse husbandry, T. Carell for a gift of isotopically labelled deoxynucleoside standards and the members of the Hajkova laboratory for discussions and revisions of the manuscript. Work in the Hajkova laboratory is supported by MRC funding (MC_US_A652_5PY70), the FP7 EpiGeneSys network and an ERC grant (ERC-CoG-648879–dynamicmodifications) to P.H. The laboratory of Y.Z. and S.P. is supported by grant 1R44GM096723-01A1. P.H. is a member of the EMBO Young Investigator Programme. P.W.S.H. is a recipient of an MRC PhD Studentship and MRC-targeted Doctoral Prize Fellowship from Imperial College London.

Author information

Authors and Affiliations

Authors

Contributions

P.H. and P.W.S.H. conceived the study; P.W.S.H. performed the experiments and analysed the data; H.G.L. carried out mouse ES cell experiments with the help of M.B. and M.R.-T.; C.E.R. generated Tet1−/− DNMT TKO mouse ES cell line with the help of H.B.; R.A. carried out LC–MS/MS experiments and analysed the data with the help of S.L.; J.T. made Aba–seq libraries with support from Y.Z.; computational analysis was carried out by P.W.S.H. with the help of Z.S., G.D., V.H., and B.L.; R.V. performed experiments; P.W.S.H. and P.H. wrote the manuscript with assistance from S.P. and B.L.

Corresponding author

Correspondence to Petra Hajkova.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks Y. Matsui and the other anonymous reviewer(s) for their contribution to the peer review of this work.

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 Figure 1 Characterization of WGBS datasets and validation of Aba–seq method.

a, Distribution of WGBS coverage for each symmetric CpG. For box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest and lowest value within 1.5 × the inter-quartile range, respectively. b, Overview of the Aba–seq method15. ce, Density heat map showing correlation between levels of 5hmC at all 2-kb windows (minimum of four symmetric CpGs) in E14 mouse ES cells as computed by TAB–seq35 (x-axis) and Aba–seq15 (y-axis) (c); TAB–seq35 (x-axis) and hMeDIP36 (y-axis) (d) or Aba–seq15 (x-axis) and hMeDIP36 (y-axis) (e). For ce, the Pearson correlation coefficient (ρ) is shown. Specific details for all Extended Data Figures regarding sample sizes and how samples were collected can be found in the ‘Statistics and reproducibility’ section.

Extended Data Figure 2 Further analysis of the levels of 5hmC in E10.5 PGCs.

a, Density heat map showing the levels of 5hmC per 2-kb window (with minimum four CpGs) for E10.5 PGCs (y-axis) and E14 mouse ES cells15 (x-axis). The Pearson correlation coefficient (ρ) is shown. b, Levels of 5hmC (ascertained using Aba–seq) at various regulatory elements in E10.5 PGCs (left) or E14 mouse ES cells15 (right). P values are based on ANOVA and Dunnett’s post hoc test. For box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest and lowest value within 1.5 × the inter-quartile range, respectively. c, Metagene plot showing the levels of 5hmC (determined using Aba–seq) (top) and the combined levels of 5mC and 5hmC (determined using WGBS) (bottom) in E10.5 PGCs across genes expressed at different levels in E10.5 PGCs. d, e, Metagene plot showing the levels of 5hmC (determined using Aba–seq) (top) and the combined levels of 5mC and 5hmC (determined using WGBS) (bottom) in E10.5 PGCs across either putative active enhancers (d) or CpG islands (e). f, Bar chart showing the levels of 5hmC at ICRs in E14 mouse ES cells as determined by TAB–seq35 (%; light green) or Aba–seq15 (read counts; dark green), or in E10.5 PGCs as determined by Aba–seq (read counts; orange).

Extended Data Figure 3 Further analysis of 5mC and 5hmC dynamics in PGCs.

a, Combined levels of 5mC and 5hmC as determined by WGBS (left) or levels of 5hmC as determined by Aba–seq (right) at various features within the uniquely mapped part of the genome in PGCs between E10.5 and E12.5. For box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest or lowest value within 1.5 × the inter-quartile range, respectively. b, The combined levels of 5mC and 5hmC (determined by WGBS) (left) or levels of 5hmC (determined by Aba–seq) (right) at various consensus repetitive elements in PGCs between E10.5 and E12.5. Asterisks indicate mean values.

Extended Data Figure 4 5hmC is targeted to newly hypomethylated regions following DNA demethylation in mouse gonadal PGCs.

See also Extended Data Fig. 5. a, Density heat map showing Pearson correlation (ρ) between levels of 5hmC for E10.5 PGC biological replicates (left), for E10.5 and E11.5 PGCs (middle), and for E10.5 and E12.5 PGCs (right). b, Mean Z-scores depicting levels of 5hmC (determined by Aba–seq) (orange) and combined levels of 5mC and 5hmC (determined by WGBS) (grey) for each stage normalized to the average level of either 5hmC or combined 5mC and 5hmC across stages. Standard error of the mean is shown but it is too small to see. cf, Density heat maps showing the correlation between the total (c, d) or relative (e, f) levels of 5hmC in E10.5 (c, e) or E11.5 (d, f) PGCs and the change in the combined levels of 5mC and 5hmC in PGCs between these two stages for all 2-kb windows with a minimum 20% combined 5mC and 5hmC in E10.5 PGCs. g, Density heat map showing the correlation between the relative levels of 5hmC in E11.5 PGCs and the combined level of 5mC and 5hmC in E11.5 PGCs for all 2-kb windows with a minimum 20% combined 5mC and 5hmC in E10.5 PGCs. h, Combined levels of 5mC and 5hmC in E10.5 and E11.5 PGCs for 2-kb windows with a minimum 20% combined 5mC and 5hmC in E10.5 PGCs that are either 1) enriched for total levels of 5hmC at either E10.5 or E11.5 (green, upper-tail adjusted Poisson P < 0.05), or 2) depleted of total 5hmC at both E10.5 and E11.5 (red, lower-tail adjusted Poisson P < 0.05). i, Density plot showing the decrease in combined levels of 5mC and 5hmC in PGCs between E10.5 and E11.5 for 2-kb windows with a minimum 20% total DNA modification in E10.5 PGCs that are either 1) enriched for total levels of 5hmC at either E10.5 or E11.5 (green, upper-tail adjusted Poisson P < 0.05), or 2) depleted of total 5hmC at both E10.5 and E11.5 (red, lower-tail adjusted Poisson P < 0.05). For all box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest or lowest value within 1.5 × the inter-quartile range, respectively. P values are based on a two-sided Wilcoxon test. Note that for density heat maps, the Spearman correlation (ρS) is shown and the red line represents the smoothed mean as determined by a generalized additive model.

Extended Data Figure 5 Suggested models implicating 5mC oxidation in DNA demethylation of gonadal PGCs.

a, A model of oxidation followed by passive dilution predicts a positive correlation between the extent to which the combined levels of 5mC and 5hmC decrease between two stages (as determined by WGBS) and the total level of 5hmC at both the stage immediately preceding and following the decrease. b, A model implicating 5mC oxidation in triggering DNA demethylation via an active mechanism predicts a positive correlation between the extent to which the combined levels of 5mC and 5hmC decrease between two stages (as determined by WGBS) and the relative levels of 5hmC in the stage immediately preceding this decrease, as further oxidation of 5hmC to 5-formylcytosine (5fC) is the rate-limiting step in the full oxidation of 5mC to 5-carboxylcytosine (5caC) (ref. 39). c, A model implicating oxidation of 5mC in safeguarding DNA hypomethylation following the major wave of DNA demethylation predicts that regions where the majority of DNA methylation has been lost between two stages (that is, those that are newly hypomethylated) will have high relative levels of 5hmC in the stage immediately after the major wave of DNA demethylation in order to remove residual methylation and/or aberrant de novo methylation. Thus, a limited correlation between the extent to which the combined levels of 5mC and 5hmC decrease between two stages (as determined by WGBS) and the relative levels of 5hmC in the stage immediately following this decrease may also be seen.

Extended Data Figure 6 Expression of TET1, TET2 and TET3 and locus-specific DNA methylation in Tet1−/− PGCs during epigenetic reprogramming.

a, Expression of Tet1 total transcript (left) or Tet1 exon 4 (right) in E12.5 Tet1−/− and wild-type PGCs. Adjusted P values (left) computed by DESeq2 and P values (right) computed by Student’s t-test. Asterisks indicate mean values. b, Representative immunostaining against the N terminus of TET1 protein in E12.5 wild-type and Tet1−/− PGCs. Scale bars represent 10 μm. c, Expression of Tet2 and Tet3 in E12.5 Tet1−/− and wild-type PGCs. Adjusted P values computed by DESeq2. Asterisks indicate mean values. d, e, Mean combined levels of 5hmC and 5mC (determined using RRBS) in female (d) or male (e) E12.5 and E14.5 Tet1−/− and wild-type PGCs for ICRs and germline gene promoters labelled as hypermethylated in E14.5 Tet1−/− PGCs. The mean DNA modification level and P values were computed using RnBeads software (see Methods). f, g, Locus-specific bisulfite sequencing of the Dazl promoter (left), the Peg3 ICR (middle) and the IG-DMR ICR (right) in E12.5 (f) and E13.5 (g) female Tet1−/− and wild-type PGCs.

Extended Data Figure 7 Promoter DNA methylation clustering analysis during germline reprogramming.

a, The combined levels of 5mC and 5hmC at promoters (ascertained using WGBS) (right), levels of 5hmC at promoters (ascertained by Aba–seq) (centre), or gene expression levels (RNA-seq data) (right) in consecutive stages of PGC development for all genes grouped by k-means clustering of the combined 5mC and 5hmC dynamics at their promoter regions. b, c, Box plots depicting the combined levels of 5mC and 5hmC at promoters (ascertained using WGBS) (left), levels of 5hmC at promoters (ascertained using Aba–seq) (centre), or gene expression levels (RNA-seq data) (right) in consecutive stages of PGC development for three clusters of genes with either low CpG promoters (b) or intermediate CpG promoters (c) grouped by k-means clustering of the combined 5mC and 5hmC dynamics at their promoter regions. For all box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest and lowest values within 1.5 × the inter-quartile range, respectively.

Extended Data Figure 8 DNA modification and expression dynamics in wild-type and Tet1−/− PGCs at retrotransposons normally activated concurrently with epigenetic reprogramming.

a, b, Combined 5mC and 5hmC dynamics in wild-type PGCs (%; WGBS; far left); relative 5hmC dynamics (Aba–seq read counts normalized to E10.5) in wild-type PGCs (centre left); the expression dynamics in either wild-type or Tet1−/− PGCs (transcripts per million (TPM); RNA-seq data; centre right); and the combined dynamics of 5mC and 5hmC in wild-type and Tet1−/− PGCs (%; RRBS; far right) for representative repetitive elements (IAPA_MM, IAPEZI and L1mdTf_II) that are significantly upregulated (adjusted P < 0.05; Sleuth) in a sex-independent manner (a), a male-specific manner (b, blue outline) or a female-specific manner (b, pink outline) between E10.5 and E14.5 in wild-type PGCs. Mean values are shown in all cases. Adjusted P values for differential repeat expression analysis between E14.5 wild-type and Tet1−/− PGCs are based on Sleuth software.

Extended Data Figure 9 Characterization of GRR gene regulation by TET1 and 5mC in PGCs and mouse ES cells.

a, CpG density at GRR gene promoters and other relevant promoters; P values are based on a two-sided Wilcoxon test. b, Mean 5hmC dynamics at GRR gene promoters and non-activated methylated and demethylated HCPs in PGCs; P values are based on a two-sided paired Wilcoxon test. c, log2(fold change) between Tet1−/− and wild-type E14.5 male (blue) or female (pink) PGCs for GRR genes and other relevant gene sets. FWER-adjusted P values are based on GSEA software (see Methods). d, log2(fold change) between Dnmt1CKO (ref. 24) and wild-type mouse PGCs (green), or between E14.5 female (pink) or male (blue) wild-type PGCs and E10.5 wild-type PGCs, for GRR genes and other relevant gene sets. FWER-adjusted P values are based on GSEA software (see Methods). e, Correlation between the difference in the combined levels of 5mC and 5hmC (%, ascertained by RRBS) (x-axis: Tet1−/− − wild type (WT)) at GRR promoters and the change in GRR gene expression (y-axis; log2(Tet1−/−/wild type)) in E12.5 (left) and E14.5 (right) Tet1−/− PGCs. Spearman’s correlation is shown. f, Representative western blot showing TET1 and lamin B protein expression in wild-type, DNMT TKO, and Tet1−/− DNMT TKO mouse ES cells. For all box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest or lowest value within 1.5 × the inter-quartile range, respectively.

Extended Data Figure 10 Epigenetic characterization of GRR gene promoters in mouse ES cells.

a, Genomic sequences centred on transcription start sites of GRR genes, non-GRR genes activated in both male and female PGCs between E10.5 and E14.5, and non-GRR methylated and demethylated HCP genes in wild-type mouse ES cells grown in serum-containing medium. Each horizontal line represents one gene; the intensity of red indicates the relative enrichment for the feature shown at the top of each column. The transcription start site and sequences 5 kb upstream and downstream of the transcription start site are shown. bf, Box plots depicting the combined levels of 5mC and 5hmC (ascertained using WGBS)30 (b); levels of 5hmC (ascertained using Aba–seq)15 (c); levels of TET1 (ChIP–seq data)21 (d); levels of RING1B (ChIP–seq data)38 (e) and levels of H2Aub (ChIP–seq data)37 (f) at the promoters of GRR genes and of other relevant gene sets in wild-type mouse ES cells grown in serum-containing media. For all box plots, the upper and lower hinges correspond to the first and third quartiles, the centre line corresponds to the median, and the maxima and minima correspond to the highest and lowest value within 1.5 × the inter-quartile range, respectively. P values are based on a two-sided Wilcoxon test. g, Metagene plot depicting median levels of H3K4me3 (ChIP–seq data)30 around the transcription start sites of GRR genes (left) and non-GRR HCP genes that are also initially methlylated and subsequently demethylated during PGC reprogramming (right) in wild-type and Tet1−/− mouse ES cells grown in serum-containing medium. P values are based on a paired two-sided Wilcoxon test for the promoter (−1 kb/+500 bp) region.

Extended Data Figure 11 Characterization of GRR gene regulation by PRC1 and 5mC in PGCs and mouse ES cells.

a, Overlap between GRR genes and genes significantly upregulated in E11.5 and/or E12.5 PRC1 CKO PGCs compared with wild-type26. P values based on hypergeometric test. b, Representative western blot showing H2Aub and H2A levels in wild-type or DNMT TKO mouse ES cells after 6 h DMSO treatment, and wild-type or DNMT TKO mouse ES cells after 6 h PRT4165 treatment. c, Classification of GRR genes on the basis of their dependency for 5mC and/or PRC1 reprogramming in mouse ES cells (see Methods).

Extended Data Figure 12 Model.

The timely and efficient activation of GRR genes, involved in the transition from PGC to gonocyte and the correct progression of gametogenesis, requires interactions between promoter CpG density, the initiation of global DNA demethylation, TET1 recruitment, and removal of PRC1-mediated repression. Both DNA demethylation-dependent (safeguarding against aberrant residual and/or de novo promoter DNA methylation) and -independent (such as the potential recruitment of OGT or other transcriptional activators to gene promoters22,28) functions of TET1 are important for GRR gene activation.

Supplementary information

Life Sciences Reporting Summary (PDF 87 kb)

Supplementary Information

This file contains Supplementary Tables 1-6, Supplementary Figure 1 and Supplementary References. (PDF 375 kb)

Supplementary Table 7

This table contains GRR genes, E10 PGC gene FPKM values and differential expression in PGCs and mESCs. (XLSX 24156 kb)

Supplementary Table 8

This table shows differential methylation analysis in PGCs. (XLSX 16215 kb)

Supplementary Table 9

This table shows differential expression of transposable elements in Tet1-KO PGCs. (XLSX 164 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hill, P., Leitch, H., Requena, C. et al. Epigenetic reprogramming enables the transition from primordial germ cell to gonocyte. Nature 555, 392–396 (2018). https://doi.org/10.1038/nature25964

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature25964

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