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:

The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells

Abstract

N6-methyladenosine (m6A) is an abundant nucleotide modification in mRNA that is required for the differentiation of mouse embryonic stem cells. However, it remains unknown whether the m6A modification controls the differentiation of normal and/or malignant myeloid hematopoietic cells. Here we show that shRNA-mediated depletion of the m6A-forming enzyme METTL3 in human hematopoietic stem/progenitor cells (HSPCs) promotes cell differentiation, coupled with reduced cell proliferation. Conversely, overexpression of wild-type METTL3, but not of a catalytically inactive form of METTL3, inhibits cell differentiation and increases cell growth. METTL3 mRNA and protein are expressed more abundantly in acute myeloid leukemia (AML) cells than in healthy HSPCs or other types of tumor cells. Furthermore, METTL3 depletion in human myeloid leukemia cell lines induces cell differentiation and apoptosis and delays leukemia progression in recipient mice in vivo. Single-nucleotide-resolution mapping of m6A coupled with ribosome profiling reveals that m6A promotes the translation of c-MYC, BCL2 and PTEN mRNAs in the human acute myeloid leukemia MOLM-13 cell line. Moreover, loss of METTL3 leads to increased levels of phosphorylated AKT, which contributes to the differentiation-promoting effects of METTL3 depletion. Overall, these results provide a rationale for the therapeutic targeting of METTL3 in myeloid leukemia.

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: m6A inhibits myeloid differentiation of HSPCs.
Figure 2: m6A promotes leukemogenesis.
Figure 3: m6A is required for maintaining the translation of target mRNAs that control cell fate.
Figure 4: m6A directly controls expression of c-MYC, BCL-2 and PTEN.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. 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  Google Scholar 

  5. Li, Z. et al. FTO plays an oncogenic role in acute myeloid leukemia as a N6-methyladenosine RNA demethylase. Cancer Cell 31, 127–141 (2017).

    Article  Google Scholar 

  6. Mauer, J. et al. Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 541, 371–375 (2017).

    Article  CAS  Google Scholar 

  7. Wang, X. et al. Corrigendum: Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature 542, 260 (2017).

    Article  CAS  Google Scholar 

  8. Lin, S., Choe, J., Du, P., Triboulet, R. & Gregory, R.I. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 62, 335–345 (2016).

    Article  CAS  Google Scholar 

  9. Bagger, F.O. et al. BloodSpot: a database of gene expression profiles and transcriptional programs for healthy and malignant haematopoiesis. Nucleic Acids Res. 44 D1, D917–D924 (2016).

    Article  CAS  Google Scholar 

  10. Śledź, P. & Jinek, M. Structural insights into the molecular mechanism of the m(6)A writer complex. eLife 5, e18434 (2016).

    Article  Google Scholar 

  11. 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  Google Scholar 

  12. Wang, X. et al. Structural basis of N6-adenosine methylation by the METTL3-METTL14 complex. Nature 534, 575–578 (2016).

    Article  CAS  Google Scholar 

  13. Shi, J. et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015).

    Article  CAS  Google Scholar 

  14. Wang, T., et al. Gene essentiality profiling reveals gene networks and synthetic lethal interactions with oncogenic Ras. Cell 168, 890–903 (2017).

    Article  CAS  Google Scholar 

  15. Linder, B. et al. Single-nucleotide-resolution mapping of m6A and m6Am throughout the transcriptome. Nat. Methods 12, 767–772 (2015).

    Article  CAS  Google Scholar 

  16. Sommer, S., Lavi, U. & Darnell, J.E. Jr . The absolute frequency of labeled N-6-methyladenosine in HeLa cell messenger RNA decreases with label time. J. Mol. Biol. 124, 487–499 (1978).

    Article  CAS  Google Scholar 

  17. Ingolia, N.T., Lareau, L.F. & Weissman, J.S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. 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  Google Scholar 

  20. Park, S.M. et al. Musashi-2 controls cell fate, lineage bias, and TGF-β signaling in HSCs. J. Exp. Med. 211, 71–87 (2014).

    Article  CAS  Google Scholar 

  21. Li, S. et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat. Med. 22, 792–799 (2016).

    Article  CAS  Google Scholar 

  22. Sarry, J.E. et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rgc-deficient mice. J. Clin. Invest. 121, 384–395 (2011).

    Article  CAS  Google Scholar 

  23. Stoneley, M. et al. c-Myc protein synthesis is initiated from the internal ribosome entry segment during apoptosis. Mol. Cell. Biol. 20, 1162–1169 (2000).

    Article  CAS  Google Scholar 

  24. Pandey, S. & Wang, E. Cells en route to apoptosis are characterized by the upregulation of c-fos, c-myc, c-jun, cdc2, and RB phosphorylation, resembling events of early cell-cycle traverse. J. Cell. Biochem. 58, 135–150 (1995).

    Article  CAS  Google Scholar 

  25. Sherrill, K.W., Byrd, M.P., Van Eden, M.E. & Lloyd, R.E. BCL-2 translation is mediated via internal ribosome entry during cell stress. J. Biol. Chem. 279, 29066–29074 (2004).

    Article  CAS  Google Scholar 

  26. Ping, X.L. et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 24, 177–189 (2014).

    Article  CAS  Google Scholar 

  27. Kharas, M.G. et al. Constitutively active AKT depletes hematopoietic stem cells and induces leukemia in mice. Blood 115, 1406–1415 (2010).

    Article  CAS  Google Scholar 

  28. Sykes, S.M. et al. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell 146, 697–708 (2011).

    Article  CAS  Google Scholar 

  29. Cui, Q. et al. m6A RNA methylation regulates the self-renewal and tumorigenesis of glioblastoma stem cells. Cell Reports 18, 2622–2634 (2017).

    Article  CAS  Google Scholar 

  30. Zhang, S., et al. m6A demethylase ALKBH5 maintains tumorigenicity of glioblastoma stem-like cells by sustaining FOXM1 expression and cell proliferation program. Cancer Cell 31, 591–606 (2017).

    Article  CAS  Google Scholar 

  31. Kwok, C.T., Marshall, A.D., Rasko, J.E. & Wong, J.J. Genetic alterations of m(6)A regulators predict poorer survival in acute myeloid leukemia. J. Hematol. Oncol. 10, 39 (2017).

    Article  Google Scholar 

  32. Patil, D.P. et al. m(6)A RNA methylation promotes XIST-mediated transcriptional repression. Nature 537, 369–373 (2016).

    Article  CAS  Google Scholar 

  33. Olarerin-George, A.O. & Jaffrey, S.R. MetaPlotR: a Perl/R pipeline for plotting metagenes of nucleotide modifications and other transcriptomic sites. Bioinformatics 33, 1563–1564 (2017).

    Article  CAS  Google Scholar 

  34. Bailey, T.L. et al. MEME SUITE: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–208 (2009).

    Article  Google Scholar 

  35. Kruse, S. et al. A novel synthesis and detection method for cap-associated adenosine modifications in mouse mRNA. Sci. Rep. 1, 126 (2011).

    Article  Google Scholar 

  36. Reid, D.W., Shenolikar, S. & Nicchitta, C.V. Simple and inexpensive ribosome profiling analysis of mRNA translation. Methods 91, 69–74 (2015).

    Article  CAS  Google Scholar 

  37. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  Google Scholar 

  38. Love, M.I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

We thank D. Bachovchin (MSKCC, New York) for the MOLM-13 constitutively expressing Cas9 cell line, and C. Vakoc (Cold Spring Harbor Laboratory, New York) for the constitutively expressing Cas9-RN2c cell line. We would like to thank the members of the Scaltriti laboratory (MSKCC, New York) for providing us with PI3K/AKT inhibitors. We thank the Weill Cornell Medicine Epigenomics Core for their assistance with sequencing. M.G.K. was supported by the US National Institutes of Health National Institute of Diabetes Digestive and Kidney Diseases Career Development Award, NIDDK NIH R01-DK101989-01A1, NCI 1R01CA193842-01, Kimmel Scholar Award, V-Scholar Award, Geoffrey Beene Award, Leukemia Lymphoma Society Career Development Award and Alex's Lemonade Stand A Award. This work was also supported by a Tri-Institutional Stem Cell Award (M.G.K. and S.R.J.), R01CA186702 (S.R.J.), T32CA062948 (B.F.P.), Ruth L. Kirschstein National Research Service Award 1F32CA22104-01 (B.F.P.), a Damon Runyon-Sohn Pediatric Cancer Fellowship Award DRSG10-14 (L.P.V.), and the American-Italian Cancer Foundation (S.Z.). The research was funded in part through the NIH/NCI Cancer Support Core Grant P30 CA08748 MGK. The RPPA core facility is funded by NCI #CA16672.

Author information

Authors and Affiliations

Authors

Contributions

L.P.V. led the project, performed experiments, analyzed data and wrote the manuscript. B.F.P., Y.C., D.N. and S.Z. performed experiments, analyzed data and edited the manuscript. G.M., T.C. and A.C. provided experimental supports. C.E.M., Y.S. and M.M. performed and analyzed MeRIP-seq experiments on patient-derived samples. J.S., C.F., M.P., F.E.G.-B., A.M., V.M.K. and M.C. provided patient samples. S.R.J. supervised the project and wrote the manuscript. M.G.K. directed the project, analyzed data and wrote the manuscript.

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

Corresponding authors

Correspondence to Samie R Jaffrey or Michael G Kharas.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures

Supplementary Figures 1–6 (PDF 1925 kb)

Life Sciences Reporting Summary (PDF 86 kb)

Supplementary Dataset

Supplementary Tables 1–13 (XLSX 9967 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vu, L., Pickering, B., Cheng, Y. et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 23, 1369–1376 (2017). https://doi.org/10.1038/nm.4416

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4416

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