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 co-transcriptionally directs the demethylation of histone H3K9me2

Abstract

A dynamic epigenome is critical for appropriate gene expression in development and health1,2,3,4,5. Central to this is the intricate process of transcription6,7,8,9,10,11, which integrates cellular signaling with chromatin changes, transcriptional machinery and modifications to messenger RNA, such as N6-methyladenosine (m6A), which is co-transcriptionally incorporated. The integration of these aspects of the dynamic epigenome, however, is not well understood mechanistically. Here we show that the repressive histone mark H3K9me2 is specifically removed by the induction of m6A-modified transcripts. We demonstrate that the methyltransferase METTL3/METTL14 regulates H3K9me2 modification. We observe a genome-wide correlation between m6A and occupancy by the H3K9me2 demethylase KDM3B, and we find that the m6A reader YTHDC1 physically interacts with and recruits KDM3B to m6A-associated chromatin regions, promoting H3K9me2 demethylation and gene expression. This study establishes a direct link between m6A and dynamic chromatin modification and provides mechanistic insight into the co-transcriptional interplay between RNA modifications and histone modifications.

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: Identification of histone modifications responding to m6A methylation.
Fig. 2: m6A methylation regulates H3K9me2 levels.
Fig. 3: Genome-wide correlation between m6A and KDM3B.
Fig. 4: RNA m6A co-transcriptionally directs KDM3B to target chromatin to demethylate H3K9me2.
Fig. 5: YTHDC1 recruits KDM3B to regulate H3K9me2.
Fig. 6: RNA m6A promotes H3K9me2-regulated gene expression.

Similar content being viewed by others

Data availability

Protein expression data were downloaded from HPM database31. The histone ChIP–seq data for H3K9me2, H3K27me3, H3K4me, H3K4me3, H3K9me, H3K9me3, H3K27ac and H3K36me3 were downloaded from the NCBI GEO database with the accession numbers GSM1314605, GSM2596866, GSM1847705, GSM2938885, GSM1003751, GSM2386847, GSM1163096 and GSM2123549, respectively, for window correlation analysis (Extended Data Fig. 1). KDM6A ChIP–seq data from GSM1207791 were downloaded for signal visualization in (Extended Data Fig. 3). RNA-seq, m6A-seq and ChIP–seq raw data have been deposited in the Gene Expression Omnibus under accession code GSE153651 and the Genome Sequence Archive in BIG Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences under accession number CRA002304. Source data are provided with this paper.

Code availability

Custom code written in Python is available on Github (https://github.com/linjian-smu/m6AMethylation-K9me2).

References

  1. Bonasio, R., Tu, S. & Reinberg, D. Molecular signals of epigenetic states. Science 330, 612–616 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Elgin, S. C. & Reuter, G. Position-effect variegation, heterochromatin formation, and gene silencing in Drosophila. Cold Spring Harb. Perspect. Biol. 5, a017780 (2013).

    PubMed  PubMed Central  Google Scholar 

  3. Allis, C. D. & Jenuwein, T. The molecular hallmarks of epigenetic control. Nat. Rev. Genet. 17, 487–500 (2016).

    CAS  PubMed  Google Scholar 

  4. Shi, Y. et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119, 941–953 (2004).

    CAS  PubMed  Google Scholar 

  5. Tsukada, Y. et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 439, 811–816 (2006).

    CAS  PubMed  Google Scholar 

  6. Rea, S. et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 406, 593–599 (2000).

    CAS  PubMed  Google Scholar 

  7. Klose, R. J., Kallin, E. M. & Zhang, Y. JmjC-domain-containing proteins and histone demethylation. Nat. Rev. Genet. 7, 715–727 (2006).

    CAS  PubMed  Google Scholar 

  8. Nicetto, D. & Zaret, K. S. Role of H3K9me3 heterochromatin in cell identity establishment and maintenance. Curr. Opin. Genet. Dev. 55, 1–10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Volpe, T. A. et al. Regulation of heterochromatic silencing and histone H3 lysine-9 methylation by RNAi. Science 297, 1833–1837 (2002).

    CAS  PubMed  Google Scholar 

  10. Reinhart, B. J. & Bartel, D. P. Small RNAs correspond to centromere heterochromatic repeats. Science 297, 1831 (2002).

    CAS  PubMed  Google Scholar 

  11. Verdel, A. et al. RNAi-mediated targeting of heterochromatin by the RITS complex. Science 303, 672–676 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, S. & Mason, C. E. The pivotal regulatory landscape of RNA modifications. Annu. Rev. Genomics Hum. Genet. 15, 127–150 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Frye, M., Harada, B. T., Behm, M. & He, C. RNA modifications modulate gene expression during development. Science 361, 1346–1349 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Liu, J. et al. A METTL3–METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat. Chem. Biol. 10, 93–95 (2014).

    CAS  PubMed  Google Scholar 

  18. Slobodin, B. et al. Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 169, e12 (2017).

    Google Scholar 

  19. Huang, H. et al. Histone H3 trimethylation at lysine 36 guides m(6)A RNA modification co-transcriptionally. Nature 567, 414–419 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, J. et al. N(6)-Methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Yang, Y., Hsu, P. J., Chen, Y. S. & Yang, Y. G. Dynamic transcriptomic m(6)A decoration: writers, erasers, readers and functions in RNA metabolism. Cell Res. 28, 616–624 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    PubMed  Google Scholar 

  24. Huang, H. et al. Recognition of RNA N(6)-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 20, 285–295 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Alarcon, C. R. et al. HNRNPA2B1 Is a mediator of m(6)A-dependent nuclear RNA processing events. Cell 162, 1299–1308 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Xiao, W. et al. Nuclear m(6)A reader YTHDC1 regulates mRNA splicing. Mol. Cell 61, 507–519 (2016).

    CAS  PubMed  Google Scholar 

  27. Abakir, A. et al. N(6)-Methyladenosine regulates the stability of RNA:DNA hybrids in human cells. Nat. Genet. 52, 48–55 (2020).

    CAS  PubMed  Google Scholar 

  28. Yang, X. et al. m(6)A promotes R-loop formation to facilitate transcription termination. Cell Res. 29, 1035–1038 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Xiao, S. et al. The RNA N(6)-methyladenosine modification landscape of human fetal tissues. Nat. Cell Biol. 21, 651–661 (2019).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  31. Kim, M. S. et al. A draft map of the human proteome. Nature 509, 575–581 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Ebata, K. T. et al. Vitamin C induces specific demethylation of H3K9me2 in mouse embryonic stem cells via Kdm3a/b. Epigenetics Chromatin 10, 36 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Chen, J. et al. H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat. Genet. 45, 34–42 (2013).

    CAS  PubMed  Google Scholar 

  34. Blaschke, K. et al. Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 500, 222–226 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    CAS  PubMed  Google Scholar 

  36. Yao, X. et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell Res. 27, 801–814 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Wan, L. et al. Scaffolding protein SPIDR/KIAA0146 connects the Bloom syndrome helicase with homologous recombination repair. Proc. Natl Acad. Sci. USA 110, 10646–10651 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Conrad, T. & Ørom, U. A. in Enhancer RNAs: Methods and Protocols (ed. Ørom, U. A.) 1–9 (Springer, 2017).

  39. Liu, T. et al. Gcn5 determines the fate of Drosophila germline stem cells through degradation of Cyclin A. FASEB J. 31, 2185–2194 (2017).

    CAS  PubMed  Google Scholar 

  40. Zeng, Y. et al. Refined RIP-seq protocol for epitranscriptome analysis with low input materials. PLoS Biol. 16, e2006092 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Sendinc, E. et al. PCIF1 catalyzes m6Am mRNA methylation to regulate gene expression. Mol. Cell 75, e9 (2019).

    Google Scholar 

  47. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    PubMed  PubMed Central  Google Scholar 

  50. Kidder, B. L., Hu, G. & Zhao, K. ChIP-Seq: technical considerations for obtaining high-quality data. Nat. Immunol. 12, 918–922 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Thorvaldsdottir, H., Robinson, J. T. & Mesirov, J. P. Integrative genomics viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    CAS  PubMed  Google Scholar 

  53. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Yen for discussion and critical reading of the manuscript. This work was supported by the National Key R&D Program of China (grant nos. 2018YFC1004103 and 2019YFA0802300), the Natural Science Foundation of China (grant nos. 31722034, 81771643, 31801234 and 31970595) and Pearl River S&T Nova Program of Guangzhou (grant no. 201806010009).

Author information

Authors and Affiliations

Authors

Contributions

Laixin Xia conceived the research. Laixin Xia and S.X. designed and supervised the project. Y.L., K.T., X.Y., R.X., J.S., X.L., M.D., M.L., M.Y., Z.W., S.L., H.Z., T.L., J.H. and Q.L. performed experiments. Linjian Xia, Z.Z., X.C. and J.C. conducted bioinformatic analysis. Laixin Xia and S.X. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Shan Xiao or Laixin Xia.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Screen for the epigenetic signals responding to RNA m6A methylation.

a, Schematic of PCR primers to identify the site-specifically integrated pcDNA5/FRT/To plasmid in Flp-in T-Rex 293 cell line. b, PCR products of reporters using SV40-F/FRT-R or BGH-F/FRT-R to verify integration status. c, Expression level of endogenous CEBPA and reporters normalized to ACTB. Error bars indicate mean ± SD of three independent experiments. d, The efficiency of METTL3 knockdown in reporters. The expression level of METTL3 was first normalized to ACTB then to siNC. Error bars indicate mean ± SD of three independent experiments. e, Consensus motif of m6A peaks on nascent RNA. Binomial distribution test. f, Pearson correlation coefficient analysis of m6A signals with H3K9me2 and H3K27me3, H3K9me, H3K9me2, H3K4me, H3K4me3, H3K27ac and H3K36me3 in nonoverlapping, nonrepetitive windows of different sizes (in kilobases) along the mouse genome. g, Heatmap showing the distribution of methylation level of m6A, H3K9me2 and H3K27me3 at m6A peaks on nascent RNA. h, Stacking bar chart showing the fraction of RNA modified by m6A in H3K9me2 or no H3K9me2 RNA groups. RNAs with similar exon number, expression level, transcript size or density of m6A motifs were randomly sampled and compared. P = 5.71e-121, 2.94e-07, 5.06e-69, 7.41e-28, 3.53e-55 from left to right, two-sided Fisher’s exact test.

Source data

Extended Data Fig. 2 RNA m6A regulates H3K9me2 methylation.

a, The mutation scheme of the METTL3-/- cell line. b, The mutation scheme of the METTL14-/- cell line. c, The mutation scheme of the FTO-/- cell line. d, Expression level of H3K9me2 and m6A methyltransferases and demethylases normalized to Actb. Error bars indicate mean ± SD of three independent experiments. eh, Western blots of H3K9me2 in METTL3, METTL14, FTO or ALKBH5 knockdown HEK293T cells. H3 was used as a sample processing control. Two unique shRNAs were used. i. LC/MS-MS of m6A/A in WT and METTL3D395A. Error bars indicate mean ± SD of three independent experiments. j, Distribution of hypermethylated H3K9me2 regions on chromatin. k, LC/MS-MS of m6A/A in FTO and ALKBH5 knockdown HEK293 cells using shRNAs. shGFP served as a control. Bar represents average of two independent experiments. l, Clustered heatmap showing the hypermethylated and hypomethylated H3K9me2 regions in shFTO (left) or shALKBH5 (right) compared with shGFP cells. m, Distribution of hypomethylated H3K9me2 regions on chromatin in shFTO (upper) or shALKBH5 (lower) cells. n, Overlap between genes with decreased H3K9me2 signals and genes with increased m6A signals in shFTO cells (left) or shALKBH5 (right) versus shGFP. P= 5.22e-09 in ALKBH5 knockdown, P = 4.41e-21 in FTO knockdown, Hypergeometric distribution test.

Source data

Extended Data Fig. 3 Genome-wide correlation between m6A and KDM3B in H3K9me2 regulation.

a, Pearson correlation of METTL3 with KDM3B protein expression level in HPM database. ACTB served as a negative control. The gray shades represent the 95% confidence intervals of the linear regression model. b, Western blot showing the expression level of SFB-METTL3 and FLAG-KDM3B and endogenous METTL3 and KDM3B. c, Overlap between KDM3B-occupied genes and genes corresponding to m6A-modified transcripts after normalization with similar exon number, expression level, transcript length, m6A motif density, or splicing sites. P=0, 0, 0, 8.23e-107, 0 from left to right, hypergeometric distribution test. d, Heatmap showing RPKM of KDM3B and KDM6A ChIP–seq relative to m6A peak center. e, Heatmap showing RPKM of KDM3B and KDM6A ChIP–seq relative to METTL3 ChIP peak center. f, Distribution of KDM3B ChIP–seq tags relative to the m6A peak center. g, Distribution of KDM6A ChIP–seq tags relative to the METTL3 peak center.

Source data

Extended Data Fig. 4 m6A-dependent localization of KDM3B to target chromatin.

a, Western blot of the endogenous KDM3B in WT and METTL3D395A cells. TUBULIN serve as a loading control. b, Distribution of KDM3B ChIP–seq signals change around the center of METTL3 ChIP peaks in WT versus METTL3D395A cells. c, d, Heatmap showing RPKM of KDM3B ChIP–seq relative to m6A peak center (c) or METTL3 ChIP peak center (d) in WT and METTL3D395A. e, KDM3B ChIP–qPCR in WT and METTL3D395A cells. Error bars indicate mean ± SD of three independent experiments. f, Near-Infrared (NIR) western blot and quantification of H3K9me2 modification levels in WT and METTL3D395A cells before and after induced expression of Flag-KDM3B. Error bars indicate mean ± SD of five independent experiments in right panel. ** P = 0.0072, two-sided Student’s t test. g, NIR western blots and quantification of H3K9me2 in WT and METTL3D395A cells before and after vitamin C treatment. In F and G, H3 was used as a loading control. Error bars indicate mean ± SD of five independent experiments in right panel, ** P = 0.005, two-sided Student’s t test.

Source data

Extended Data Fig. 5 YTHDC1 recruits KDM3B to regulate H3K9me2.

a, Co-IP of SFB-KDM3B with MYC tagged m6A readers YTHDC1, YTHDF2 and HNRNPA2B1 in HEK293T cells. b, Fluorescence immunostaining of endogenous KDM3B and YTHDC1 in mES or HEK293T. Scale bar: 2 μM. c, Western blot of the H3K9me2 modification level after YTHDC1 knockdown by two siRNAs in HEK293T. The decrease of YTHDC1 were also shown. H3 served as sample processing control. d, ChIP–qPCR of KDM3B in shGFP and shYTHDC1 mES cells. Enrichment of ChIP versus input chromatin was normalized against spike-in drosophila chromatin. Error bars indicate mean ± SD of three independent experiments.

Source data

Extended Data Fig. 6 The relationship between gene expression, m6A peak density and H3K9me2 abundance.

a, Heatmap showing the relationship between gene expression, m6A peak density and H3K9me2 abundance in m6A modified transcripts with increased H3K9me2 signals after METTL3 mutation, other m6A modified transcripts without H3K9me2 increase, and non-m6A modified transcripts in METTL3D395A versus WT cells.

Supplementary information

Reporting Summary

Supplementary Table 1

List of primers used in this study.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Xia, L., Tan, K. et al. N6-Methyladenosine co-transcriptionally directs the demethylation of histone H3K9me2. Nat Genet 52, 870–877 (2020). https://doi.org/10.1038/s41588-020-0677-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-020-0677-3

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing