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:

N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells

Abstract

N6-methyladenosine (m6A) has been identified as the most abundant internal modification of messenger RNA in eukaryotes1. m6A modification is involved in cell fate determination in yeast2,3 and embryo development in plants4,5. Its mammalian function remains unknown but thousands of mammalian mRNAs and long non-coding RNAs (lncRNAs) show m6A modification6,7 and m6A demethylases are required for mammalian energy homeostasis and fertility8,9. We identify two proteins, the putative m6A MTase, methyltransferase-like 3 (Mettl3; ref. 10), and a related but uncharacterized protein Mettl14, that function synergistically to control m6A formation in mammalian cells. Knockdown of Mettl3 and Mettl14 in mouse embryonic stem cells (mESCs) led to similar phenotypes, characterized by lack of m6A RNA methylation and lost self-renewal capability. A large number of transcripts, including many encoding developmental regulators, exhibit m6A methylation inversely correlated with mRNA stability and gene expression. The human antigen R (HuR) and microRNA pathways were linked to these effects. This gene regulatory mechanism operating in mESCs through m6A methylation is required to keep mESCs at their ground state and may be relevant to thousands of mRNAs and lncRNAs in various cell types.

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: Mettl3 and Mettl14 are required for m6A formation in vitro and in vivo.
Figure 2: Mettl3 and Mettl14 interact and regulate each other’s stability.
Figure 3: Mettl3 or Mettl14 KD mESCs lose self-renewal capability.
Figure 4: m6A modification regulates mRNA stability.
Figure 5: The HuR-microRNA pathway functions in m6A-methylation-mediated RNA stability.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Jia, G., Fu, Y. & He, C. Reversible RNA adenosine methylation in biological regulation. Trends Genet. 29, 108–115 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Clancy, M. J., Shambaugh, M. E., Timpte, C. S. & Bokar, J. A. Induction of sporulation in Saccharomyces cerevisiae leads to the formation of N6-methyladenosine in mRNA: a potential mechanism for the activity of the IME4 gene. Nucleic Acids Res. 30, 4509–4518 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Agarwala, S. D., Blitzblau, H. G., Hochwagen, A. & Fink, G. R. RNA methylation by the MIS complex regulates a cell fate decision in yeast. PLoS Genet. 8, e1002732 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhong, S. et al. MTA is an Arabidopsis messenger RNA adenosine methylase and interacts with a homolog of a sex-specific splicing factor. Plant Cell 20, 1278–1288 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bodi, Z. et al. Adenosine methylation in Arabidopsis mRNA is associated with the 3’ end and reduced levels cause developmental defects. Front. Plant Sci. 3, 1–10 (2012).

    Article  Google Scholar 

  6. Meyer, K. D. et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3’ UTRs and near stop codons. Cell 149, 1635–1646 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Zheng, G. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Jia, G. et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bokar, J. A., Shambaugh, M. E., Polayes, D., Matera, A. G. & Rottman, F. M. Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase. RNA 3, 1233–1247 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bujnicki, J. M., Feder, M., Radlinska, M. & Blumenthal, R. M. Structure prediction and phylogenetic analysis of a functionally diverse family of proteins homologous to the MT-A70 subunit of the human mRNA:m(6)A methyltransferase. J. Mol. Evol. 55, 431–444 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Petrossian, T. C. & Clarke, S. G. Uncovering the human methyltransferasome. Mol. Cell. Proteom. 10, M110.000976-1-12 (2011).

    Article  Google Scholar 

  13. Ku, M. et al. Genomewide analysis of PRC1 and PRC2 occupancy identifies two classes of bivalent domains. PLoS Genet. 4, e1000242 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  14. van den Berg, D. L. et al. An Oct4-centered protein interaction network in embryonic stem cells. Cell Stem Cell 6, 369–381 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Srikantan, S., Tominaga, K. & Gorospe, M. Functional interplay between RNA-binding protein HuR and microRNAs. Curr. Protein Peptide Sci. 13, 372–379 (2012).

    Article  CAS  Google Scholar 

  16. Kundu, P., Fabian, M. R., Sonenberg, N., Bhattacharyya, S. N. & Filipowicz, W. HuR protein attenuates miRNA-mediated repression by promoting miRISC dissociation from the target RNA. Nucleic Acids Res. 40, 5088–5100 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kishore, S. et al. A quantitative analysis of CLIP methods for identifying binding sites of RNA-binding proteins. Nat. Methods 8, 559–564 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Lebedeva, S. et al. Transcriptome-wide analysis of regulatory interactions of the RNA-binding protein HuR. Mol. Cell 43, 340–352 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Le, M. T. et al. Conserved regulation of p53 network dosage by microRNA-125b occurs through evolving miRNA-target gene pairs. PLoS Genet. 7, e1002242 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lu, L., Katsaros, D., de la Longrais, I. A., Sochirca, O. & Yu, H. Hypermethylation of let-7a-3 in epithelial ovarian cancer is associated with low insulin-like growth factor-II expression and favorable prognosis. Cancer Res. 67, 10117–10122 (2007).

    Article  CAS  PubMed  Google Scholar 

  21. Leung, A. K. et al. Genome-wide identification of Ago2 binding sites from mouse embryonic stem cells with and without mature microRNAs. Nat. Struct. Mol. Biol. 18, 237–244 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhao, J., Sun, B. K., Erwin, J. A., Song, J. J. & Lee, J. T. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science 322, 750–756 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Harper, J. E., Miceli, S. M., Roberts, R. J. & Manley, J. L. Sequence specificity of the human mRNA N6-adenosine methylase in vitro. Nucleic Acids Res. 18, 5735–5741 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhao, J. et al. Genome-wide identification of polycomb-associated RNAs by RIP-seq. Mol. Cell 40, 939–953 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, Y. et al. Reprogramming of mouse and human somatic cells by high-performance engineered factors. EMBO Rep. 12, 373–378 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  29. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  31. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Robinson, J. T. et al. Integrative genomics viewer. Nature Publishing Group 29, 24–26 (2011).

    CAS  Google Scholar 

  33. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Gen. Biol. 9, R137 (2008).

    Article  Google Scholar 

  34. Durinck, S., Spellman, P. & Birney, E. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protocol. 4, 1184–1191 (2009).

    Article  CAS  Google Scholar 

  35. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocol. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  36. Smyth, G. in Bioinformatics and Computational Biology Solutions Using R and Bioconductor (eds Gentle- man, R., Carey, V., Dudoit, S., Irizarry, R. & Huber, W.) 397–420 (Springer, 2005).

    Book  Google Scholar 

  37. Mukherjee, N. et al. Integrative regulatory mapping indicates that the RNA-binding protein HuR couples pre-mRNA processing and mRNA stability. Mol. Cell 43, 327–339 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Wilkinson and all members of his laboratory for many valuable discussions; the Mass Spectrometry, Metabolomics, and Proteomics Facility at the University of Illinois at Chicago for measuring m6A/A ratios; S. S. Govindarajan from the Analytical Genomics core facility at the Sanford Burnham Medical Institute (Lake Nona) for performing the microarray analysis; and P. Ordoukhanian and S. R. Head from the Scripps Research Institute for high-throughput sequencing analysis. This work is supported by a CIRM Training Grant TG2-01162 (Y.W.), an AACR-Aflac, Career Development Award for Pediatric Cancer Research 12-20-10-ZHAO (J.C.Z.), the Ontario Research Fund - Global Leader (Round 2) and Natural Sciences and Engineering Research Council (NSERC; Z.Z.), and an NSERC Canada Graduate Scholarship and Ontario Graduate Scholarship (Y.L.). M.D.P. is an American Cancer Society Research Scholar (RSG-11-224-01-DMC). Z.Z. would like to dedicate this paper to the memory of S.Z.

Author information

Authors and Affiliations

Authors

Contributions

Y.W. generated Mettl3 and Mettl14 KD mESC lines, and using these lines, designed, performed and analysed most experiments, except meRIP, RIPs and EMSAs. meRIP, RIPs and EMSAs were performed by Y.W. and J.C.Z. Y.L. performed all bioinformatics analysis under the supervision of Z.Z. J.I.T. and M.D.P. purified luciferase, Mettl3 and/or Mettl14 proteins from baculovirus-infected Sf9 insect cells. J.C.Z. formulated, analysed and directed the study and wrote the paper.

Corresponding author

Correspondence to Jing Crystal Zhao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Mettl3 and Mettl14 are required for m6A formation in vitro and in vivo.

A. Schematic drawing showing predicted MTase domains of mouse Mettl3 (Accession: NP_062695.2) and Mettl14 (Accession: NP_964000.2) proteins. Numbers represent amino acid numbers. B. RT-qPCR (left) and western blot (right) showing Mettl3 (left panel) and Mettl14 (right panel) expression in mESC clones stably expressing shRNAs targeting each. GapDH serves as a loading control in the western blot. Scr: scramble; shM3, shRNA against Mettl3; shM14, shRNA against Mettl14. The original gel is shown in Supplementary Fig. 5. C. Ethidium bromide staining (left panel) and m6A immunoblot (right panel) of DNA-free, rRNA-free Poly(A)+ RNA from Mettl3 kd, Mettl14 kd, and control cells. D. Measurement of percentage of m6A/A ratio by MS. E. Flag, Mettl3 or Mettl14 western immunoblots of proteins purified from lysates of HEK293 cells overexpressing flag-tagged Mettl3, Mettl14, or luciferase using M2 beads. Luc, luciferase; M3, Mettl3; M14, Mettl14. The original gel is shown in Supplementary Fig. 5. F. CPM counts of RNA probes extracted after an in vitro methylation assay. G. CPM counts of excised m6A spots from digested RNA probes used in the methylation assay. Error bars from panels B and F-G represent means ± SEM from 3 separate experiments, expect M3 in panel G where n = 7. One-tailed Student’s t-test, *P<0.05,**P<0.01,***P<0.001,****P<0.0001 vs. scramble control. Scr: scramble control.

Supplementary Figure 2 Mettl3 and Mettl14 interact and regulate each other’s stability.

A. meRIP-seq analysis of a Sox17 transcript showing loss of m6A methylation in both Mettl3 kd and Mettl14 kd cells. Yellow highlighting, peak location. Scr: scramble; shM3, shRNA against Mettl3; shM14, shRNA against Mettl14. B. Venn diagram showing overlap of Mettl3 and Mettl14 targets. C. Mettl3 or Mettl14 western immunoblots of endogenous proteins co-IP’d using antibodies against Mettl3 or Mettl14, or IgG in mESC lysates. M3, Mettl3; M14, Mettl14. The original gel is shown in Supplementary Fig. 5. D. Western blot analysis of Mettl3 and Mettl14 protein levels in Mettl3 kd and Mettl14 kd mESCs, respectively. The original gel is shown in Supplementary Fig. 5. E. RT-qPCR analysis of Mettl3 and Mettl14 RNA levels in Mettl3 kd and Mettl14 kd mESCs, respectively. F. GO analysis of 4395 shared Mettl3 and Mettl14 targets. Error bars from panel E represent means ± SEM from 3 separate experiments. One-tailed Student’s t-test, **P<0.01,***P<0.001 vs. scramble control. Scr: scramble.

Supplementary Figure 3 Mettl3 or Mettl14 knockdown mESCs lose self-renewal capability.

A. Phase contrast microscopy showing colony morphology of kd versus control mESC cells. Scr: scramble; shM3, shRNA against Mettl3; shM14, shRNA against Mettl14. B. Growth curve assessing cell proliferation kinetics of kd versus control cells. P values are generated by two-way ANOVA. C. Quantification of AP-positive colonies. D. Heat map analysis based on microarray comparison of gene expression in kd and control cells. E. RT-qPCR analysis of pluripotency (left) and differentiation (right) genes in kd versus control cells. F. GSEA analysis on enrichment of developmental regulators (left) and pluripotency-related genes (right) in Mettl3 kd versus control cells. A False Discovery Rate (FDR) of <0.178 was calculated for bivalent genes and FDR<0 for pluripotency-related genes. Note that FDR<0.25 is statistically significant for GSEA analysis: www.broadinstitute.org/gsea/doc/GSEAUserGuideFrame.html. Scale bars, 100 μm. Error bars from panels B-C and E represent mean± SEM from 3 separate experiments. One-tailed Student’s t-test, *P<0.05,**P<0.01,***P<0.001,****P<0.0001 vs. scramble control. Scr: scramble.

Supplementary Figure 4 m6A modification regulates mRNA stability.

A. Enrichment of up- or down-regulated Mettl3 and Mettl14 targets in kd versus control cells. Arrows indicate up- or down regulation of gene expression. Scr: scramble; shM3, shRNA against Mettl3; shM14, shRNA against Mettl14. B. Comparison of cumulative transcript abundance in cells treated with Actinomycin D for 0, 4, and or 8 hrs by microarray analysis. RNAs from 0 hr serves as base-line. KS test, p<2.2e−16 comparing the changes in relative RNA levels from 4 to 8 hrs between Mettl targets and non-targets in both Mettl3 kd and Mettl14 kd cells. C. Enrichment of bivalent and pluripotency (Oct4 PPI) genes in shared Mettl3 and Mettl14 targets. D. meRIP-qPCR of specific bivalent and pluripotency genes (grey area). E. Enrichment of Mettl3 and Mettl14 targets showing increased RNA stability in kd cells among bivalent and pluripotency genes. F. Measurement of a percentage of m6A/A ratio by MS in undifferentiated (day 0), day6, and day 12 differentiated mESCs. G. RT-qPCR of Mettl3, Mettl14, and Rex1 expression in undifferentiated and differentiated mESCs. H. meRIP-qPCR of specific bivalent (gray area) and pluripotency genes in mESCs during differentiation. Error bars from panels D and G-H represent means ± SEM from 3 separate experiments. One-tailed Student’s t-test, *P<0.05,**P<0.01,***P<0.001,****P<0.0001 vs. scramble control. Scr: scramble.

Supplementary Figure 5 The HuR-microRNA pathway functions in m6A methylation-mediated RNA stability.

A. Enrichment of genes showing increased RNA stability following Mettl3 or Mettl14 kd among all shared Mettl3 and Mettl14 targets and among targets with and without canonical HuR binding sites in their 3’-UTR regions. M3, Mettl3; M14, Mettl14. B. Left panel, denaturing PolyAcrylamide Gel Electrophoresis (PAGE) showing the size of fragmented, DNAse-treated, rRNA-free mRNAs extracted from scramble and kd cells; mid-panel, non-denaturing PAGE showing differential binding of HuR to RNAs probes from left panel. Scr: scramble control, shM14, shRNA against Mettl14, shM3, shRNA against Mettl3. NP: no protein added. Right panel, non-denaturing PAGE showing differential binding of HuR to RNA probes enlisted in C. C. RNA probes with the canonical UUUUU HuR binding site located either next to A (0A) or m6A (0m6A) or separated by a 12-nucleotide spacer (12A and 12m6A). D. HuR RIP-qPCR of Pou5f1, Igfbp3, and Otx2 from kd versus control cells. E. RT-qPCR of Pou5f1, Igfbp3, and Otx2 in Actinomycin D-treated kd cells. In the case of Igfbp3, cells are also treated with and without siRNA targeting HuR. P values are generated using two-way ANOVA. F. RT-qPCR of HuR, Igfbp3, Otx2, and Pou5f1 in mESC with depleted HuR. shHuR#1-3: shRNAs against HuR. G. Argonaute 2 (Ago2) RIP–qPCR of Igfbp3 and Pou5f1 RNAs from kd versus control cells. H. Enrichment of genes showing increased RNA stability and expression among shared Mettl3 and Mettl14 targets. Targets are classified by whether they display Ago2 binding sites. I. Model: In wildtype mESCs, Mettl3 and Mettl14 methylate RNA synergistically and m6A methylation on some transcripts, particularly those encoding developmental regulators, blocks HuR binding, resulting in transcript destabilization. In Mettl3 and Mettl14 kd cells, loss of m6A allows HuR-mRNA interaction and attenuation of microRNA targeting, enhancing stability of transcripts especially those encoding developmental regulators, and promoting loss of the mESC ground state. A potentially methylated A is shown in red. Error bars from panels D-G represent means ± SEM from 3 separate experiments. One-tailed Student’s t-test, *P<0.05,**p<0.01,****P<0.0001 vs. scramble control. Scr: scramble.

Supplementary information

Supplementary Information

Supplementary Information (PDF 740 kb)

Supplementary Table 1

Supplementary Information (XLSX 1190 kb)

Supplementary Table 2

Supplementary Information (XLSX 3597 kb)

Supplementary Table 3

Supplementary Information (XLSX 33 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, Y., Li, Y., Toth, J. et al. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nat Cell Biol 16, 191–198 (2014). https://doi.org/10.1038/ncb2902

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2902

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