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:

m6A RNA methylation orchestrates transcriptional dormancy during paused pluripotency

Abstract

Embryos across metazoan lineages can enter reversible states of developmental pausing, or diapause, in response to adverse environmental conditions. The molecular mechanisms that underlie this remarkable dormant state remain largely unknown. Here we show that N6-methyladenosine (m6A) RNA methylation by Mettl3 is required for developmental pausing in mouse blastocysts and embryonic stem (ES) cells. Mettl3 enforces transcriptional dormancy through two interconnected mechanisms: (1) it promotes global mRNA destabilization and (2) it suppresses global nascent transcription by destabilizing the mRNA of the transcriptional amplifier and oncogene N-Myc, which we identify as a crucial anti-pausing factor. Knockdown of N-Myc rescues pausing in Mettl3−/− ES cells, and forced demethylation and stabilization of Mycn mRNA in paused wild-type ES cells largely recapitulates the transcriptional defects of Mettl3−/− ES cells. These findings uncover Mettl3 as a key orchestrator of the crosstalk between transcriptomic and epitranscriptomic regulation during developmental pausing, with implications for dormancy in adult stem cells and cancer.

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

Access options

Buy this article

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

Fig. 1: The m6A methyltransferase Mettl3 is essential for paused pluripotency.
Fig. 2: Mettl3 regulates hypotranscription in paused pluripotency.
Fig. 3: The methyltransferase activity of Mettl3 sustains paused pluripotency.
Fig. 4: Mettl3 promotes RNA destabilization during pausing.
Fig. 5: The transcription factor N-Myc is a key mediator of the pausing defects of Mettl3−/− ES cells.
Fig. 6: Mettl3 regulates pausing through m6A-mediated destabilization of Mycn mRNA.

Similar content being viewed by others

Data availability

Sequencing data have been deposited into the NCBI Gene Expression Omnibus (GEO) repository (http://ncbi.nlm.nih.gov/geo) under accession number GSE202848. Mass spectrometry data have been deposited into the Metabolights database (https://www.ebi.ac.uk/metabolights) under the identifier MTBLS8041. Published RNA-seq and ChIP–seq data used in this study are available under the accession numbers E-MTAB-2958 (early mouse embryos), E-MTAB-3386 (Myc/Mycn DKO ES cells) and GSE11431 (N-Myc ChIP). Examples of FACS gating have been deposited into the Figshare repository (https://doi.org/10.6084/m9.figshare.23551986). Mouse and human reference genomes (mm10 and hg19, respectively) were downloaded from the UCSC browser (https://genome.ucsc.edu/). All other data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

Code availability

Code supporting this study are available at a dedicated GitHub repository (https://github.com/EvelyneCollignon/Mettl3_pausing) and at Zenodo (https://doi.org/10.5281/zenodo.8068381).

References

  1. Renfree, M. B. & Fenelon, J. C. The enigma of embryonic diapause. Development 144, 3199–3210 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. van der Weijden, V. A. & Bulut-Karslioglu, A. Molecular regulation of paused pluripotency in early mammalian embryos and stem cells. Front. Cell Dev. Biol. 9, 2039 (2021).

    Google Scholar 

  3. Fenelon, J. C. & Renfree, M. B. The history of the discovery of embryonic diapause in mammals. Biol. Reprod. 99, 242–251 (2018).

    Article  PubMed  Google Scholar 

  4. Boroviak, T. et al. Lineage-specific profiling delineates the emergence and progression of naive pluripotency in mammalian embryogenesis. Dev. Cell 35, 366–382 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bulut-Karslioglu, A. et al. Inhibition of mTOR induces a paused pluripotent state. Nature 540, 119–123 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bulut-Karslioglu, A. et al. The transcriptionally permissive chromatin state of embryonic stem cells is acutely tuned to translational output. Cell Stem Cell 22, 369–383 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Scognamiglio, R. et al. Myc depletion induces a pluripotent dormant state mimicking diapause. Cell 164, 668–680 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jiang, X. et al. The role of m6A modification in the biological functions and diseases. Signal Transduct. Target Ther. 6, 74 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wang, P., Doxtader, K. A. & Nam, Y. Structural basis for cooperative function of Mettl3 and Mettl14 methyltransferases. Mol. Cell 63, 306–317 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Batista, P. J. et al. m6A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell 15, 707–719 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Geula, S. et al. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 347, 1002–1006 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Wang, Y. et al. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat. Cell Biol. 16, 191–198 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lasman, L. et al. Context-dependent compensation between functional Ythdf m6A reader proteins. Genes Dev. 34, 1373–1391 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu, J. et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chelmicki, T. et al. m6A RNA methylation regulates the fate of endogenous retroviruses. Nature 591, 312–316 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Wei, J. et al. FTO mediates LINE1 m6A demethylation and chromatin regulation in mESCs and mouse development. Science 376, 968–973 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Xu, W. et al. METTL3 regulates heterochromatin in mouse embryonic stem cells. Nature 591, 317–321 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Percharde, M., Bulut-Karslioglu, A. & Ramalho-Santos, M. Hypertranscription in development, stem cells, and regeneration. Dev. Cell 40, 9–21 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Hussein, A. M. et al. Metabolic control over mTOR-dependent diapause-like state. Dev. Cell 52, 236–250 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sousa, M. I., Correia, B., Rodrigues, A. S. & Ramalho-Santos, J. Metabolic characterization of a paused-like pluripotent state. Biochim. Biophys. Acta Gen. Subj. 1864, 129612 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Sorci, M. et al. METTL3 regulates WTAP protein homeostasis. Cell Death Dis. 9, 796 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Barbieri, I. et al. Promoter-bound METTL3 maintains myeloid leukaemia by m6A-dependent translation control. Nature 552, 126–131 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bertero, A. et al. The SMAD2/3 interactome reveals that TGFβ controls m6A mRNA methylation in pluripotency. Nature 555, 256–259 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhou, K. I. et al. Regulation of co-transcriptional pre-mRNA splicing by m6A through the low-complexity protein hnRNPG. Mol. Cell 76, 70–81 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Meyer, K. D. et al. 5′ UTR m6A promotes cap-independent translation. Cell 163, 999–1010 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gaidatzis, D., Burger, L., Florescu, M. & Stadler, M. B. Analysis of intronic and exonic reads in RNA-seq data characterizes transcriptional and post-transcriptional regulation. Nat. Biotechnol. 33, 722–729 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Kim, J., Chu, J., Shen, X., Wang, J. & Orkin, S. H. An extended transcriptional network for pluripotency of embryonic stem cells. Cell 132, 1049–1061 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Kress, T. R., Sabò, A. & Amati, B. MYC: connecting selective transcriptional control to global RNA production. Nat. Rev. Cancer 15, 593–607 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Percharde, M., Wong, P. & Ramalho-Santos, M. Global hypertranscription in the mouse embryonic germline. Cell Rep. 19, 1987–1996 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, X. et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    Article  PubMed  Google Scholar 

  32. Wang, X. et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xiao, W. et al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol. Cell 61, 507–519 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xia, Z. et al. Epitranscriptomic editing of the RNA N6-methyladenosine modification by dCasRx conjugated methyltransferase and demethylase. Nucleic Acids Res. 49, 7361–7374 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Zhao, B. S. et al. m6A-dependent maternal mRNA clearance facilitates zebrafish maternal-to-zygotic transition. Nature 542, 475–478 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Aguilo, F. et al. Coordination of m6A mRNA methylation and gene transcription by ZFP217 regulates pluripotency and reprogramming. Cell Stem Cell 17, 689–704 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. He, B. et al. Blastocyst activation engenders transcriptome reprogram affecting X-chromosome reactivation and inflammatory trigger of implantation. Proc. Natl Acad. Sci. USA 116, 16621–16630 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Huang, H. et al. Histone H3 trimethylation at lysine 36 guides m6A RNA modification co-transcriptionally. Nature 567, 414–419 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sun, H. L. et al. Stabilization of ERK-phosphorylated METTL3 by USP5 increases m6A methylation. Mol. Cell 80, 633–647 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Schöller, E. et al. Interactions, localization, and phosphorylation of the m6A generating METTL3–METTL14–WTAP complex. RNA 24, 499–512 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Laplante, M. & Sabatini, D. M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang, P. et al. mTOR is necessary for proper satellite cell activity and skeletal muscle regeneration. Biochem. Biophys. Res. Commun. 463, 102–108 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hu, J. K. H. et al. An FAK-YAP-mTOR signaling axis regulates stem cell-based tissue renewal in mice. Cell Stem Cell 21, 91–106 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rehman, S. K. et al. Colorectal cancer cells enter a diapause-like DTP state to survive chemotherapy. Cell 184, 226–242 (2021).

    Article  CAS  PubMed  Google Scholar 

  47. Dhimolea, E. et al. An embryonic diapause-like adaptation with suppressed Myc activity enables tumor treatment persistence. Cancer Cell 39, 240–256 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Liu, Y., Azizian, N. G., Sullivan, D. K. & Li, Y. mTOR inhibition attenuates chemosensitivity through the induction of chemotherapy resistant persisters. Nat. Commun. 13, 7047 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Yankova, E. et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature 593, 597–601 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Jeschke, J. et al. Downregulation of the FTO m6A RNA demethylase promotes EMT-mediated progression of epithelial tumors and sensitivity to Wnt inhibitors. Nat. Cancer 2, 611–628 (2021).

    Article  CAS  PubMed  Google Scholar 

  52. Nichols, J., Evans, E. P. & Smith, A. G. Establishment of germ-line-competent embryonic stem (ES) cells using differentiation inhibiting activity. Development 110, 1341–1348 (1990).

    Article  CAS  PubMed  Google Scholar 

  53. Handyside, A. H., O’Neill, G. T., Jones, M. & Hooper, M. L. Use of BRL-conditioned medium in combination with feeder layers to isolate a diploid embryonal stem cell line. Rouxs Arch. Dev. Biol. 198, 48–56 (1989).

    Article  PubMed  Google Scholar 

  54. Ran, F. A. et al. Genome engineering using the CRISPR–Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Macrae, T. A. & Ramalho-Santos, M. The deubiquitinase Usp9x regulates PRC2-mediated chromatin reprogramming during mouse development. Nat. Commun. 12, 1865 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Ross, R., Cao, X., Yu, N. & Limbach, P. A. Sequence mapping of transfer RNA chemical modifications by liquid chromatography tandem mass spectrometry. Methods 107, 73–78 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Delatte, B. et al. Transcriptome-wide distribution and function of RNA hydroxymethylcytosine. Science 351, 282–285 (2016).

    Article  CAS  PubMed  Google Scholar 

  58. DiTroia, S. P. et al. Maternal vitamin C regulates reprogramming of DNA methylation and germline development. Nature 573, 271–275 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Tang, Y. et al. m6A-Atlas: a comprehensive knowledgebase for unraveling the N6-methyladenosine (m6A) epitranscriptome. Nucleic Acids Res. 49, D134–D143 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Castellanos-Rubio, A. et al. A novel RT–QPCR-based assay for the relative quantification of residue specific m6A RNA methylation. Sci. Rep. 9, 4220 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Olazagoitia-Garmendia, A. & Castellanos-Rubio, A. Relative quantification of residue-specific m6A RNA methylation using m6A-RT–QPCR. Methods Mol. Biol. 2298, 185–195 (2021).

    Article  CAS  PubMed  Google Scholar 

  62. Neumann, T. et al. Quantification of experimentally induced nucleotide conversions in high-throughput sequencing datasets. BMC Bioinformatics 20, 258 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  63. DI Tommaso, P. et al. Nextflow enables reproducible computational workflows. Nat. Biotechnol. 35, 316–319 (2017).

    Article  PubMed  Google Scholar 

  64. Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 38, 276–278 (2020).

    Article  CAS  PubMed  Google Scholar 

  65. Kurtzer, G. M., Sochat, V. & Bauer, M. W. Singularity: scientific containers for mobility of compute. PLoS ONE 12, e0177459 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Sadowski, H. B. & Gilman, M. Z. Cell-free activation of a DNA-binding protein by epidermal growth factor. Nature 362, 79–83 (1993).

    Article  CAS  PubMed  Google Scholar 

  67. Méndez, J. et al. Human origin recognition complex large subunit is degraded by ubiquitin-mediated proteolysis after initiation of DNA replication. Mol. Cell 9, 481–491 (2002).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Santos Laboratory, D. Schramek, A. Bulut-Karslioglu, T. Macrae, J. Jeschke and F. Fuks for feedback on the manuscript; members of the Hanna Laboratory for providing cells; members of the UCSF Center for Advanced Technology and the LTRI Sequencing Core for next-generation sequencing; staff at the LTRI Flow Cytometry Facilities, A. Bulut-Karslioglu and S. Biechele for advice on diapause; M. Percharde and T. Macrae for bioinformatics guidance; and staff at the TCP Transgenic Core for providing timed pregnant animals and the TCP Animal Resources for colony management. E.C. was supported by a fellowship from the Belgian American Educational Foundation. P.A.L. is funded by the US National Institutes of Health (NIH, GM58843). This work was supported by grant R01GM113014 from the NIH, a Canada 150 Research Chair in Developmental Epigenetics, the Great Gulf Homes Charitable Foundation, and project grants 165935 and 178094 from the Canadian Institutes of Health Research (to M.R.-S.).

Author information

Authors and Affiliations

Authors

Contributions

E.C. and M.R.-S. conceived the project and designed experiments. E.C. performed the majority of the experiments and interpreted data. B.C. performed EU flow cytometry and embryo imaging. G.F. generated knockout ES cells and induced mouse hormonal diapause. J.F.-R. performed lentiviral infection and chromatin shearing. E.C., S.-B.M. and S.A.M. performed SLAM–seq. P.A.L. and R.L.R. performed mass spectrometry. M.R.-S. supervised the project. E.C. and M.R.-S. wrote the manuscript with feedback from all authors.

Corresponding authors

Correspondence to Evelyne Collignon or Miguel Ramalho-Santos.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Jacob Hanna, Baoming Qin, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Dissection of paused pluripotency in Mettl3−/− models.

a. m6A increase in paused ESCs was validated in an independent mass spectrometry experiment. Levels relative to control (Ctrl) for each replicate are shown (n = 3 biological replicates). b. Validation of Rptor and Rictor knockdown by RT-qPCR in paused ESCs grown in FBS/LIF/2i (n = 4 biological replicates). c. mTOR inhibition by Rptor and Rictor knockdown induces a paused phenotype with reduced cell proliferation and total RNA levels (n = 4 biological replicates). d. Dot blot showing an increase in m6A levels in ESCs upon knockdown of Rptor and Rictor. Levels of m6A are normalized to RNA loading control (methylene blue staining, n = 4 biological replicates). e. Design of Mettl3−/− ESCs models used in this study. f. Validation of Mettl3−/− ESCs, in control and pausing conditions, by western blot (representative of 3 biological replicates). g. Validation of Mettl3−/− #2–4 ESCs, in control and pausing conditions, by western blot (representative of 2 biological replicates). h. Mettl3−/− #2–4 ESCs also fail to suppress proliferation in paused conditions (n = 3 biological replicates). i. Mettl3-knockout mutant model in mice (Mettl3TCP−/−) and genotyping by PCR (left). Example of PCR genotyping of embryos resulting from Mettl3TCP+/− crossing, representative of all genotyping performed in this study [n(Mettl3TCP+/+) = 87, n(Mettl3TCP+/+) = 132, n(Mettl3TCP+/+) = 46]. +/+: wildtype, +/-: heterozygous, -/-: knockout. j. Validation of Mettl3TCP−/− in embryos by immunofluorescence. Representative staining images are shown. Number of embryos (n) as indicated. Scale bars = 30 µm. Data are mean ± SD (a-d) or mean ± SEM (h). P-values (as indicated on figure) by one-way ANOVA with Dunnett’s multiple comparison tests (a-c), two-tailed ratio paired Student’s t-tests (d), and linear regression test with interaction (h).

Source data

Extended Data Fig. 2 Paused Mettl3−/− ESCs acquire a distinct gene expression profile.

a. Quantification of total RNA per cell in Mettl3+/+ #2 and Mettl3−/− #2–4 ESCs, in control and paused conditions. Data are mean ± SD, n = 5 biological replicates. b. Decrease in nascent RNA per cell, as measured by EU incorporation with nuclear signal quantification, in wildtype ex vivo paused or hormonally diapaused blastocysts, compared to control E3.5 embryos. Number of embryos (n) as indicated. Scale bars = 30 µm. c. Strategy for RNA-seq with cell number-normalization using ERCC spike-in RNAs. d. Quantification of the number of expressed genes in Mettl3+/+ and Mettl3−/− ESCs, in control and paused conditions. Expressed genes are further defined as having high expression (log2 normalized reads > 5, n = 3 biological replicates). e. Number of differentially expressed genes (fold-change > 1.5 and adjusted P < 0.05) upon pausing, in Mettl3+/+ and Mettl3−/− ESCs. f. PCA plot for all expressed genes across all samples, showing across PC1 that Mettl3+/+ ESCs acquire a more divergent expression profile upon pausing than Mettl3−/− ESCs, relative to respective control condition. g-h. Gene expression changes (log2 fold-changes) of gene sets selected from the ‘GO biological processes’ (g) and ‘hallmarks’ (h) collections, showing incomplete downregulation in paused Mettl3−/− ESCs. i. Western blot of total and phosphorylated mTOR, and of the downstream targets of mTORC1 (Ulk1, 4Ebp1 and S6K1) (left, representative of 4 biological replicates). Quantification of phosphorylated levels, normalized to total levels, show no significant change in paused ESCs between Mettl3+/+ and Mettl3−/− (right). Data are mean ± SD (b, d, g-i). P-values (as indicated on figure) by two-way ANOVA with Dunnett’s multiple comparison tests (a, i), one-way ANOVA with Dunnett’s multiple comparison tests (b), two-tailed unpaired Student’s t-tests (d, g-h).

Source data

Extended Data Fig. 3 Mapping m6A distribution in the transcriptome of paused ESCs.

a. Strategy for MeRIP-seq in ESCs with cell number-normalization (CNN) using human cell spiking. b. Validation of the CNN strategy for the MeRIP-seq. By mixing different ratios of human cells to ESCs (1, 2 or 4%), we simulated global changes in methylation. Spiking normalization allows capture of these differences, as shown here by MeRIP-qPCR for 3 methylated mRNAs (NeuroD1, Nr5a2, Sox1). Data are mean ± SD, n = 5 biological replicates with levels relative to 2% spike in each replicate. c. The specificity of the m6A capture was tested by spiking poly(A) RNA from ESCs with exogenous RNAs before performing MeRIP-qPCR. Data are mean ± SD, n = 3 biological replicates. P-values (as indicated on figure) by two-tailed unpaired Student’s t-tests. d. Examples of gene track views of MeRIP-seq, for mRNAs of pluripotency factors previously shown to be methylated in ESCs. e. Motif analysis performed with DREME in a 100 bp window surrounding MeRIP peak summits identifies several motifs corresponding to the consensus ‘DRACH’ m6A motif (where D = A, G or U; H = A, C or U). f. Distribution of differential m6A peaks, according to the type of structural element within the transcript. g. Examples of gene track views of MeRIP-seq, for mRNAs with significant hypermethylation (Tial1, Ptrf) or hypomethylation (Cenpt) in pausing of ESCs. h. GSEA of m6A changes in paused ESCs relative to control ESCs, using the ‘hallmarks’ collection. No single pathway is significantly enriched based on m6A changes (representative pathways are shown). P-values (as indicated on figure) by two-sided pre-ranked gene set enrichment analysis with Benjamini-Hochberg FDR correction.

Source data

Extended Data Fig. 4 Mapping the chromatin distribution of Mettl3 in paused ESCs.

a. m6A machinery (writers Mettl3, Mettl14 and Wtap; and erasers Fto and Alkbh5) in control (Ctrl) and paused ESCs by western blot in whole cell extracts (representative of 3 biological replicates). b. Increase of Mettl3 levels in chromatin extracts upon induction of paused pluripotency, measured by cell number-normalized (CNN) western blot (representative of 3 biological replicates). c. Strategy for Mettl3 ChIP-seq in ESCs with CNN approach using human cell spiking. d. Heatmap of the top 5000 most variable Mettl3 peaks by ChIP-seq across all samples, showing higher levels in paused ESCs (n = 2 biological replicates per group). e. Density plot of the average levels of Mettl3 binding in the TSS of all genes by ChIP-seq, separated by expression and methylation status, in control and paused ESCs. Mettl3 binding is highest in the TSS of expressed genes with a methylated transcript, and in paused ESCs. Number of genes (n) as indicated. Data as mean normalized Mettl3 level (n = 2 biological replicates per group). f. Examples of gene track views showing increased average levels (fold-change > 1.5) of m6A and Mettl3, by MeRIP-seq and Mettl3 ChIP-seq, respectively.

Source data

Extended Data Fig. 5 Capturing Mettl3-dependent changes in RNA stability in paused ESCs.

a. Strategy for RNA stability analysis based on intronic and exonic reads. b-c. Examples of genes with different (Slc16a1, Six4) and similar (Mtor, Gapdh) intronic and exonic mRNA fold-changes between Mettl3/ and Mettl3+/+ ESCs based on RNA-seq data (b) and validation of stability changes by actinomycin D stability assay (c). N = 3 biological replicates, t1/2: half-life. d. Linear regression of log2 total conversion counts (relative to time 0 h), as measured by SLAM-seq, showing an increase in transcriptome stability in paused Mettl3/ ESCs. e. Changes in RNA expression in paused Mettl3/ ESCs based on RNA-seq data (fold-change > 1.5) are associated with changes in RNA half-life in paused Mettl3−/− ESCs. Data are mean ± SD (b) or mean ± SEM (c). P-values (as indicated on figure) by two-tailed paired Student’s t-tests (b), linear regression test with interaction (c) and one-way ANOVA (e). Boxes in the box plots define the interquartile range (IQR) split by the median, with whiskers extending to the most extreme values within 1.5 × IQR beyond the box.

Source data

Extended Data Fig. 6 Screening for candidate anti-pausing factors.

a. Quantification of the number of expressed genes in Mettl3+/+ and Mettl3/ ESCs based on intronic RNA-seq, in control and paused conditions. Expressed genes are further defined as having high expression (log2 normalized reads > 5, n = 3 biological replicates). b. Heatmap of gene expression based on intronic reads for all genes expressed in Mettl3+/+ or Mettl3/ ESCs (left) with average expression per sample (right, scored as median z-scores of all genes), showing defective hypotranscription in paused Mettl3/ ESCs. c. Identification of putative anti-pausing factors kept in check by m6A methylation and thereby destabilization of their transcript in paused pluripotency, based on RNA-seq, MeRIP-seq and SLAM-seq data in ESCs (see Methods for details). d. Expression levels (log2 cpm) of the Myc factors in diapaused embryos (left, data from Boroviak et al.) and paused ESCs (right). Horizontal bars represent the mean, with 3 biological replicates per group, except for diapaused embryos which has 2 replicates. e. mTOR inhibition by dual knockdown of Rptor and Rictor reduces Mycn expression measured by RT-qPCR in ESCs in FBS/LIF/2i medium (n = 4 biological replicates). Data are mean ± SD (a, b, e). P-values (as indicated on figure) by two-way ANOVA with Tukey’s multiple comparisons test (a), two-tailed Student’s t-tests (b), and one-way ANOVA with Dunnett’s multiple comparison tests (e).

Source data

Extended Data Fig. 7 Regulation of Myc family members and downstream targets by Mettl3 in paused pluripotency.

a. Quantification of N-Myc protein levels, showing increased expression in Mettl3/ ESCs, as shown in Fig. 5c. N = 4 biological replicates. b. Representative western blot of N-Myc protein levels, showing increased expression in Mettl3/− #2–4 ESCs. N = 2 biological replicates. c. Increased expression of N-Myc in Mettl3−/− ESCs grown in FBS/LIF/2i (compared to Mettl3+/+ ESCs) measured by RT-qPCR (n = 3 biological replicates). Levels are normalized to control Mettl3+/+ ESCs grown in FBS/LIF, as shown in Fig. 5b. d. Mycn expression in Mettl3−/− ESCs is restored to levels comparable to Mettl3+/+ ESCs by transfecting a catalytically active form of Mettl3, and not its inactive mutant form (RT-qPCR, n = 4 biological replicates). e. Validation of Mycn knockdown by RT-qPCR in paused ESCs grown in FBS/LIF (left) or FBS/LIF/2i (right). f-g. Blocking of Myc signaling by Mycn knockdown (e, in FBS/LIF/2i) or chemical inhibitor 10058-F4 (f, in FBS/LIF) partially restores the pausing phenotype in paused Mettl3/ ESCs in terms of cell proliferation (left, n = 3 biological replicates) and total RNA levels per cell (right, n = 4 and 5 biological replicates). h. Treatment with 10058-F4 partially restores the expression of Myc target genes in paused Mettl3/ ESCs (RT-qPCR, n = 5 biological replicates). Data are mean ± SD (a, c-h), or mean ± SEM (g left). P-values (as indicated on figure) by two-tailed unpaired Student’s t-tests (a, d, f), by two-way ANOVA with Tukey’s multiple comparisons test (c) or Dunnett’s multiple comparison tests (d-e, h), one-way ANOVA with Dunnett’s multiple comparison tests (f-g).

Source data

Extended Data Fig. 8 m6A-dependent regulation of Mycn mRNA stability.

a. Knockout of Ythdf2 and triple knockout of Ythdf1-3 (TKO) phenocopy the knockout of Mettl3 in paused ESCs, with increased total RNA levels per cell (left, n = 4 biological replicates) and proliferation (right, n = 4 biological replicates) compared to wildtype (WT) ESCs. b. Increased expression of N-Myc in paused Ythdf2−/− and TKO ESCs measured by RT-qPCR (n = 4 biological replicates, relative to paused WT). c. Validation of m6A changes in Mettl3+/+ and Mettl3/ ESCs by m6A-qPCR (n = 6 biological replicates). d. Mettl3 and Ythdf2 binding of the Mycn transcript, measured by RIP-qPCR in 3 biological replicates. NeuroD1 and Sox2 were used as positive controls, and Actb and Gapdh were used as negative controls. e-f. Mycn is the only Myc family member regulated at the RNA stability level by Mettl3, as evidenced by analysis of exonic and intronic mRNA fold-changes (left, n = 3 biological replicates per group) and SLAM-seq analysis of RNA half-life (right, with half-lives derived from 2 independent time courses). g. Nascent RNA capture by EU incorporation shows minimal changes in nascent transcription for Myc factors in Mettl3/ ESCs (n = 5 biological replicates). h. Increased Mycn mRNA stability in paused Ythdf2−/− ESCs compared to paused Mettl3+/+ ESCs, as measured by an actinomycin D stability assay (n = 3 biological replicates). t1/2: half-life. Data are mean ± SD (a-e, g) or mean ± SEM (h). P-values (as indicated on figure) by one-way ANOVA with Dunnett’s multiple comparison tests (a-c), two-tailed Student’s t-tests (d-e), two-way ANOVA with Dunnett’s multiple comparison tests (g), and linear regression test with interaction (h).

Source data

Extended Data Fig. 9 Targeted m6A demethylation controls expression of Mycn in paused ESCs.

a. Model of lentiviral dCasRx epitranscriptomic editor with the m6A demethylase ALKBH5 and single guide RNA. b. Validation of the expression of the dCasRx-ALKBH5 fusion by western blot in parental (non-infected) ESCs, infected ESCs, and 2 infected clones. Clone 5 was used for all experiments (representative of 2 biological replicates). c. Guide RNAs (gRNAs) transfected for non-targeting control and Mycn-targeting conditions. d. Changes in m6A using the dCasRx-ALKBH5 editor in paused ESCs. Guides #2 and #3 significantly reduce m6A in Mycn transcripts, as measured by m6A-qPCR, and were selected for all subsequent experiments. N = 7 biological replicates. NT: non-targeting. e. Dot blot showing that the global increase of m6A in paused ESCs is not affected by the dCasRx-ALKBH5 editor, with Mettl3−/− ESCs as negative control. Representative of 3 biological replicates. MB: methylene blue. f. Quantification of N-Myc protein levels, showing increased expression with the dCasRx-ALKBH5 editor targeting Mycn in paused ESCs, with representative blot shown in Fig. 6e. N = 3 biological replicates. g. Demethylation of Mycn increases the total RNA levels per cell in paused ESCs (n = 6 biological replicates). Data are mean ± SD (d, f-g) and P-values (as indicated on figure) by one-way ANOVA with Dunnett’s multiple comparison tests (d, f-g).

Source data

Extended Data Fig. 10 Transcriptional changes by RNA-seq upon m6A demethylation of Mycn in paused ESCs.

a. Mycn expression is increased following targeting with the m6A demethylase ALKBH5 based on exonic reads, but not intronic reads, which is consistent with post-transcriptional regulation (n = 4 biological replicates per condition). b. A global increase in transcripts levels is measured following Mycn mRNA demethylation using both exonic and intronic reads, which is consistent with globally elevated nascent transcription. c. Representative pathways of the GSEA of gene expression changes upon demethylation of Mycn mRNA in paused ESCs using the ‘hallmarks’ collection. d-e. Genes upregulated upon demethylation of Mycn mRNA are enriched for N-Myc targets, as identified in ESCs by ChIP by Chen et al.36. GSEA using a random set of 100 N-Myc targets (c). Venn diagram showing a significant overlap between genes upregulated in paused Mettl3/ ESCs, genes upregulated following Mycn demethylation, and N-Myc ChIP targets. Data are mean ± SD (a). P-values (as indicated on figure) by two-way ANOVA with Dunnett’s multiple comparison tests (a), two-sided pre-ranked gene set enrichment analysis with Benjamini-Hochberg FDR correction (c, d), and one-sided simulation using hypergeometric distributions (e).

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables 1–7

Supplementary Table 1: Differential expression in paused ES cells with Mettl3 KO from RNA-seq data. N = 3 biological replicates per group. Supplementary Table 2: GSEA using RNA-seq data. Expression data presented in Supplementary Table 1 (n = 3 biological replicates per group). Supplementary Table 3: Differential methylation analysis from MeRIP–seq data. N = 3 biological replicates per group. Supplementary Table 4: RNA decay analysis from SLAM–seq data. Half-lives were measured using four time points, with samples collected over two independent experiments. Supplementary Table 5: Top 100 ranked m6A anti-pausing candidates in paused pluripotency. Supplementary Table 6: Differential expression with Mycn mRNA demethylation in paused ES cells from RNA-seq data. N = 4 biological replicates per group. Supplementary Table 7: List of primers and antibodies.

Source data

Source Data Fig. 1

Numerical source data related to Fig. 1.

Source Data Fig. 1

Image source data related to Fig. 1.

Source Data Fig. 2

Numerical source data related to Fig. 2.

Source Data Fig. 3

Numerical source data related to Fig. 3.

Source Data Fig. 3

Image source date related to Fig. 3.

Source Data Fig. 4

Numerical source data related to Fig. 4.

Source Data Fig. 5

Numerical source data related to Fig. 5.

Source Data Fig. 5

Image source data related to Fig. 5c,d.

Source Data Fig. 6

Numerical source data related to Fig. 6.

Source Data Extended Data Fig. 1

Numerical source data related to Extended Data Fig. 1.

Source Data Extended Data Fig. 1

Image source data related Extended Data Fig.1c,f,g,i.

Source Data Extended Data Fig. 2

Numerical source data related to Extended Data Fig. 2.

Source Data Extended Data Fig. 2

Image source data related to Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Numerical source data related to Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Image source data related to Extended Data Fig.4a, b.

Source Data Extended Data Fig. 5

Numerical source data related to Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Numerical source data related to Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Numerical source data related to Extended Data Fig. 7.

Source Data Extended Data Fig. 7

Image source data related to Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Numerical source data related to Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Numerical source data related to Extended Data Fig. 9.

Source Data Extended Data Fig. 9

Image source data related to Extended Data Fig. 9b,e.

Source Data Extended Data Fig. 10

Numerical source data related to Extended Data Fig. 10.

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

Collignon, E., Cho, B., Furlan, G. et al. m6A RNA methylation orchestrates transcriptional dormancy during paused pluripotency. Nat Cell Biol 25, 1279–1289 (2023). https://doi.org/10.1038/s41556-023-01212-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01212-x

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