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 modification controls the innate immune response to infection by targeting type I interferons

A Publisher Correction to this article was published on 11 January 2019

This article has been updated

Abstract

N6-methyladenosine (m6A) is the most common mRNA modification. Recent studies have revealed that depletion of m6A machinery leads to alterations in the propagation of diverse viruses. These effects were proposed to be mediated through dysregulated methylation of viral RNA. Here we show that following viral infection or stimulation of cells with an inactivated virus, deletion of the m6A ‘writer’ METTL3 or ‘reader’ YTHDF2 led to an increase in the induction of interferon-stimulated genes. Consequently, propagation of different viruses was suppressed in an interferon-signaling-dependent manner. Significantly, the mRNA of IFNB, the gene encoding the main cytokine that drives the type I interferon response, was m6A modified and was stabilized following repression of METTL3 or YTHDF2. Furthermore, we show that m6A-mediated regulation of interferon genes was conserved in mice. Together, our findings uncover the role m6A serves as a negative regulator of interferon response by dictating the fast turnover of interferon mRNAs and consequently facilitating viral propagation.

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: m6A ‘writers’ and ‘readers’ are upregulated during HCMV infection and required for viral growth.
Fig. 2: Inhibition of HCMV growth in m6A-deficient cells is driven by an enhanced type I interferon response.
Fig. 3: ISG-enhanced expression in METTL3-depleted cells is independent of viral gene expression.
Fig. 4: IFNB mRNA is m6A modified and is more stable in METTL3-depleted cells.
Fig. 5: Depletion of m6A machinery proteins leads to an elevation in IFNB levels following infection with diverse viruses.
Fig. 6: Type I interferon m6A methylation and destabilization are conserved in mouse.

Similar content being viewed by others

Data availability

All RNA-seq data sets generated in this manuscript have been deposited in the GEO under accession number GSE114019. Full images of immunoblots presented in this study have been deposited to Mendeley Data and are available at https://doi.org/10.17632/3zb63b6ssj.1. All other data are available from the corresponding author upon reasonable request.

Change history

  • 11 January 2019

    In the version of this article initially published, the penultimate sentence of the abstract included a typographical error (‘cxgenes’). The correct word is ‘genes’. The error has been corrected in the HTML and PDF version of the article.

References

  1. 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 

  2. 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 

  3. Ke, S. et al. m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover. Genes Dev. 31, 990–1006 (2017).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  5. Wang, X. et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  7. Du, H. et al. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4–NOT deadenylase complex. Nat. Commun. 7, 12626 (2016).

    Article  CAS  Google Scholar 

  8. Meyer, K. D. & Jaffrey, S. R. Rethinking m6A readers, writers, and erasers. Annu. Rev. Cell Dev. Biol. 33, 319–342 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  10. Zhao, B. S., Roundtree, I. A. & He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 18, 31–42 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  12. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  14. 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  Google Scholar 

  15. Schwartz, S. et al. High-resolution mapping reveals a conserved, widespread, dynamic mRNA methylation program in yeast meiosis. Cell 155, 1409–1421 (2013).

    Article  CAS  Google Scholar 

  16. Fustin, J.-M. et al. RNA-methylation-dependent RNA processing controls the speed of the circadian clock. Cell 155, 793–806 (2013).

    Article  CAS  Google Scholar 

  17. Xiang, Y. et al. RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature 543, 573–576 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. Lence, T. et al. m6A modulates neuronal functions and sex determination in Drosophila. Nature 540, 242–247 (2016).

    Article  CAS  Google Scholar 

  20. Zhang, C. et al. m6A modulates haematopoietic stem and progenitor cell specification. Nature 549, 273–276 (2017).

    Article  CAS  Google Scholar 

  21. Yoon, K.-J. et al. Temporal control of mammalian cortical neurogenesis by m6A methylation. Cell 171, 877–889.e17 (2017).

    Article  CAS  Google Scholar 

  22. Li, H.-B. et al. m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 548, 338–342 (2017).

    Article  CAS  Google Scholar 

  23. Tong, J. et al. m6A mRNA methylation sustains Treg suppressive functions. Cell Res. 28, 253–256 (2018).

    Article  Google Scholar 

  24. Kennedy, E. M., Courtney, D. G., Tsai, K. & Cullen, B. R. Viral epitranscriptomics. J. Virol. 91, e02263–16 (2017).

    Article  CAS  Google Scholar 

  25. Courtney, D. G. et al. Epitranscriptomic enhancement of influenza A virus gene expression and replication. Cell Host Microbe 22, 377–386.e5 (2017).

    Article  CAS  Google Scholar 

  26. Gokhale, N. S. et al. N6-methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe 20, 654–665 (2016).

    Article  CAS  Google Scholar 

  27. Hesser, C., Karijolich, J., Dominissini, D., He, C. & Glaunsinger, B. A. N6-methyladenosine modification and the YTHDF2 reader protein play cell type specific roles in lytic viral gene expression during Kaposi’s sarcoma-associated herpesvirus infection. PLoS Pathog. 14, e1006995 (2018).

    Article  Google Scholar 

  28. Kennedy, E. M. et al. Posttranscriptional m(6)A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe 19, 675–685 (2016).

    Article  CAS  Google Scholar 

  29. Lichinchi, G. et al. Dynamics of human and viral RNA methylation during Zika virus infection. Cell Host Microbe 20, 666–673 (2016).

    Article  CAS  Google Scholar 

  30. Lichinchi, G. et al. Dynamics of the human and viral m6A RNA methylomes during HIV-1 infection of T cells. Nat. Microbiol. 1, 16011 (2016).

    Article  CAS  Google Scholar 

  31. Tan, B. et al. Viral and cellular N6-methyladenosine and N6,2’-O-dimethyladenosine epitranscriptomes in the KSHV life cycle. Nat. Microbiol. 3, 108–120 (2018).

    Article  CAS  Google Scholar 

  32. Tirumuru, N. et al. N6-methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag protein expression. eLife 5, e15528 (2016).

    Article  Google Scholar 

  33. Tsai, K., Courtney, D. G. & Cullen, B. R. Addition of m6A to SV40 late mRNAs enhances viral structural gene expression and replication. PLoS Pathog. 14, e1006919 (2018).

    Article  Google Scholar 

  34. Ye, F., Chen, E. R. & Nilsen, T. W. Kaposi’s sarcoma-associated herpesvirus utilizes and manipulates RNA N6-adenosine methylation to promote lytic replication. J. Virol. 91, e00466–17 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Tirosh, O. et al. The transcription and translation landscapes during human cytomegalovirus infection reveal novel host–pathogen interactions. PLoS Pathog. 11, e1005288 (2015).

    Article  Google Scholar 

  36. O’Connor, C. M., Vanicek, J. & Murphy, E. A. Host microRNA regulation of human cytomegalovirus immediate early protein translation promotes viral latency. J. Virol. 88, 5524–5532 (2014).

    Article  Google Scholar 

  37. Lin, Q. et al. Enantioselective synthesis of Janus kinase inhibitor INCB018424 via an organocatalytic aza-Michael reaction. Org. Lett. 11, 1999–2002 (2009).

    Article  CAS  Google Scholar 

  38. Karikó, K., Buckstein, M., Ni, H. & Weissman, D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23, 165–175 (2005).

    Article  Google Scholar 

  39. Durbin, A. F., Wang, C., Marcotrigiano, J. & Gehrke, L. RNAs containing modified nucleotides fail to trigger RIG-I conformational changes for innate immune signaling. mBio 7, e00833–16 (2016).

    Article  CAS  Google Scholar 

  40. Ivashkiv, L. B. & Donlin, L. T. Regulation of type I interferon responses. Nat. Rev. Immunol. 14, 36–49 (2014).

    Article  CAS  Google Scholar 

  41. Schoggins, J. W. & Rice, C. M. Interferon-stimulated genes and their antiviral effector functions. Curr. Opin. Virol. 1, 519–525 (2011).

    Article  CAS  Google Scholar 

  42. Schwartz, S. et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5’ sites. Cell Rep. 8, 284–296 (2014).

    Article  CAS  Google Scholar 

  43. Fuchs, S. Y. Hope and fear for interferon: the receptor-centric outlook on the future of interferon therapy. J. Interf. Cytokine Res. 33, 211–225 (2013).

    Article  CAS  Google Scholar 

  44. Yoshimura, A., Naka, T. & Kubo, M. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 7, 454–465 (2007).

    Article  CAS  Google Scholar 

  45. Sarasin-Filipowicz, M. et al. Alpha interferon induces long-lasting refractoriness of JAK–STAT signaling in the mouse liver through induction of USP18/UBP43. Mol. Cell. Biol. 29, 4841–4851 (2009).

    Article  CAS  Google Scholar 

  46. Gracias, D. T. et al. The microRNA miR-155 controls CD8+ T cell responses by regulating interferon signaling. Nat. Immunol. 14, 593–602 (2013).

    Article  CAS  Google Scholar 

  47. Versteeg, G. A. & García-Sastre, A. Viral tricks to grid-lock the type I interferon system. Curr. Opin. Microbiol. 13, 508–516 (2010).

    Article  CAS  Google Scholar 

  48. Zheng, Q., Hou, J., Zhou, Y., Li, Z. & Cao, X. The RNA helicase DDX46 inhibits innate immunity by entrapping m6A-demethylated antiviral transcripts in the nucleus. Nat. Immunol. 18, 1094–1103 (2017).

    Article  CAS  Google Scholar 

  49. Shi, H. et al. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 27, 315–328 (2017).

    Article  CAS  Google Scholar 

  50. Li, A. et al. Cytoplasmic m6A reader YTHDF3 promotes mRNA translation. Cell Res. 27, 444–447 (2017).

    Article  CAS  Google Scholar 

  51. Achdout, H. et al. Enhanced recognition of human NK receptors after influenza virus infection. J. Immunol. 171, 915–923 (2003).

    Article  CAS  Google Scholar 

  52. Wang, X. et al. Murine cytomegalovirus abortively infects human dendritic cells, leading to expression and presentation of virally vectored genes. J. Virol. 77, 7182–7192 (2003).

    Article  CAS  Google Scholar 

  53. Meningher, T. et al. Relationships between A(H1N1)pdm09 influenza infection and infections with other respiratory viruses. Influenza Other Respir. Viruses 8, 422–430 (2014).

    Article  Google Scholar 

  54. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  Google Scholar 

  55. Straussman, R. et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012).

    Article  CAS  Google Scholar 

  56. 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).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  58. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Schwartz and the rest of the Stern-Ginossar laboratory members for discussions and critical reading of the manuscript. This research was supported by the European Research Council starting grant (StG-2014-638142), the EU-FP7-PEOPLE Career integration grant, the ICORE (Chromatin and RNA Gene Regulation) and the Israeli Science Foundation (1073/14). N.S.-G. is incumbent of the Skirball career development chair in new scientists.

Author information

Authors and Affiliations

Authors

Contributions

R.W., E.G., L.L., J.H.H., S.S. and N.S.-G. conceived experiments and interpreted data. L.L., S.G. and J.H.H. generated and characterized the gene-deficient mice. R.W., E.G., M.S., S.G., C.S., A.N., J.T.-S., N.F. and M.M. executed experiments and analysis. V.T.K.L.-T. and M.T. provided critical reagents and advice. R.W., E.G. and N.S.-G. wrote the manuscript with contribution from all other authors.

Corresponding author

Correspondence to Noam Stern-Ginossar.

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.

Integrated supplementary information

Supplementary Figure 1 m6A machinery is elevated along HCMV infection and is important for its propagation.

a, mRNA and translation levels of genes encoding for m6A machinery along HCMV infection, as measured by RNA-seq (red) and ribosome profiling (green)35. b, Immunoblot analysis of m6A machinery proteins in cells expressing sgRNAs targeting control gene (WT) or the various m6A machinery genes (indicated on the left) in fibroblasts. GAPDH was used as a loading control. The gel images were cropped to present only relevant proteins. c, Quantification of m6A machinery proteins levels from the immunoblot analysis in b normalized to the levels of GAPDH. d, Fluorescent microscopy of GFP signal in WT fibroblasts infected with supernatant from infected cells in which m6A machinery genes were depleted (indicated at the bottom). e, Immunoblot analysis of HCMV Immediate-early protein (IE1-pp72) (left panel) and fluorescent microscopy of GFP signal (right panel), at 24 hpi in ALKBH5-depleted and control cells. GAPDH was used as a loading control. The gel images were cropped to present only relevant proteins. f, Quantification of viral protein levels from the immunoblot analysis in Fig.1e normalized to the levels of GAPDH. Data are representative of three (d) or two (e) independent experiments.

Supplementary Figure 2 Differences in ISG expression between METTL3-depleted and control cells is abolished by ruxolitinib and does not stem from changes in their stability.

a, ISG relative expression, as measured by RNA-seq, in METTL3-depleted cells versus control cells at 28 hpi, treated or untreated with ruxolitinib. Expression levels of each transcript were normalized to a scale of 0 to 1. ISGs showing significant difference (FDR < 0.01) between control and METTL3-depleted cells are presented. b-e, METTL3-depleted and control cells were treated with actinomycin D at 22 hpi and collected for RNA-seq at 0, 2 and 4 hours post treatment. The mRNA decay of several ISGs that showed enhanced expression in METTL3-depleted cells are presented (n = 2 for each time point). Values represent the mean of RNA-seq replicates and error bars show s.d. RPKM, reads per kilobase of transcript per million mapped reads. f, Quantification of protein levels from the immunoblot analysis in Fig.4a normalized to the levels of GAPDH.

Supplementary Figure 3 IFNB mRNA is m6A-modified and its levels are higher in YTHDF2-depleted cells than in control cells.

a, Specificity of m6A signal on IFNB transcript in immunoprecipitated (IP) samples compared to input (POI, Peak Over Input) and to median coverage across the gene (POM, Peak Over Median), in METTL3-depleted (n = 3) and control cells (n = 3). Thick line, median; box boundaries, 25% and 75% percentiles; whiskers, 1.5-fold interquartile range. b, IFNB mRNA and c, protein levels in YTHDF2-depleted and control cells at indicated time points post infection, measured by qRT-PCR and ELISA, respectively. 18S ribosomal RNA was used as a normalizing gene in qRT-PCR. Dots, measurements; bars, mean of three technical (b) or cell culture (c) replicates. The P values were calculated using a two-sided Student’s t-test. n.d., not detected. d, Nascent RNA was labeled for 2 h with 5-Ethynyluridine (EU). EU was washed out and RNA was extracted at the indicated time points. The relative remaining EU-labeled mRNA abundance, normalized to GAPDH, was analyzed by qRT-PCR for IFNB and USP42 that was used as control. Values represent the mean of three technical replicates and error bars show s.d. The P values were calculated using a two-sided Student’s t-test. e, IFNB gene (5′UTR, coding sequence and 3′UTR) was cloned into a plasmid in its wild-type (WT) version and in a mutant version (MUT), in which three putative m6A-modified adenosines were mutated to guanosines (labeled in red). f, Immunoblot analysis of METTL3 in THP1 cells expressing sgRNAs targeting control gene (WT) or METTL3. GAPDH was used as a loading control. The gel images were cropped to present only relevant proteins. g, Quantification of METTL3 levels from the immunoblot analysis in f normalized to the levels of GAPDH. Data (b-d) are representative of two independent experiments.

Supplementary Figure 4 m6A-mediated IFN regulation is conserved in mouse.

a, RNA-seq of input RNA and m6A immunoprecipitated RNA from mouse dendritic cells treated with lipopolysaccharide (LPS) for 3 and 6 h is presented for Ifna14. b, Immunoblot analysis of m6A machinery proteins in MEFs expressing sgRNAs targeting control gene (WT) or the various m6A machinery genes (indicated on the left). GAPDH was used as a loading control. The gel images were cropped to present only relevant proteins. c, Quantification of m6A machinery protein levels from the immunoblot analysis in b normalized to the levels of GAPDH.

Supplementary Figure 5 Construction of the Ythdf3–/– mouse.

a, Ythdf3–/– mice were generated via one-cell embryo CRISPR/Cas9 injection. sgRNA targeting Ythdf3 exon3 was used. The mutated Ythdf3 gene contains an out of frame 14bp deletion, which leads to the production of a stop codon. b, Immunoblot analysis of YTHDF3 protein expression in MEFs extracted from Ythdf3+/+, Ythdf3+/- and Ythdf3–/– embryos. HSP90 was used as a loading control. The gel images were cropped to present only relevant proteins.

Supplementary information

Supplementary Figures 1-5

Reporting Summary

Supplementary Table 1

Measurement of cell viability in control cells and cells depleted of m6A machinery proteins, before and 96 h after infection with HCMV

Supplementary Table 2

Dataset of 21 putative m6A sites in HCMV transcripts

Supplementary Table 3

Dataset of 7,093 putative m6A sites in human transcripts, obtained following infection with HCMV

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Winkler, R., Gillis, E., Lasman, L. et al. m6A modification controls the innate immune response to infection by targeting type I interferons. Nat Immunol 20, 173–182 (2019). https://doi.org/10.1038/s41590-018-0275-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0275-z

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