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:

The RNA helicase DDX46 inhibits innate immunity by entrapping m6A-demethylated antiviral transcripts in the nucleus

A Corrigendum to this article was published on 16 November 2017

This article has been updated

Abstract

DEAD-box (DDX) helicases are vital for the recognition of RNA and metabolism and are critical for the initiation of antiviral innate immunity. Modification of RNA is involved in many biological processes; however, its role in antiviral innate immunity has remained unclear. Here we found that nuclear DDX member DDX46 inhibited the production of type I interferons after viral infection. DDX46 bound Mavs, Traf3 and Traf6 transcripts (which encode signaling molecules involved in antiviral responses) via their conserved CCGGUU element. After viral infection, DDX46 recruited ALKBH5, an 'eraser' of the RNA modification N6-methyladenosine (m6A), via DDX46's DEAD helicase domain to demethylate those m6A-modified antiviral transcripts. It consequently enforced their retention in the nucleus and therefore prevented their translation and inhibited interferon production. DDX46 also suppressed antiviral innate immunity in vivo. Thus, DDX46 inhibits antiviral innate responses by entrapping selected antiviral transcripts in the nucleus by erasing their m6A modification, a modification normally required for export from the nucleus and translation.

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: DDX46 promotes VSV replication by suppressing VSV-triggered production of type I interferons in macrophages.
Figure 2: iCLIP-seq analysis of DDX46-bound RNA from VSV-infected macrophages.
Figure 3: DDX46 increases the retention of antiviral mRNA in the nucleus and decreases the expression of the protein encoded.
Figure 4: DDX46 recruits ALKBH5 to inhibit VSV-triggered production of type I interferons.
Figure 5: DDX46 affects the m6A modification of transcripts via recruitment of ALKBH5 after viral infection.
Figure 6: In vivo knockdown of DDX46 enhances the innate antiviral response.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 20 September 2017

    In the version of this article initially published, the label 'siDDX46' in the key to Figure 5g is incorrect. That label should read 'siALKBH5'. The error has been corrected in the HTML and PDF versions of this article.

References

  1. Schlee, M. & Hartmann, G. Discriminating self from non-self in nucleic acid sensing. Nat. Rev. Immunol. 16, 566–580 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gack, M.U. et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature 446, 916–920 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Cao, X. Self-regulation and cross-regulation of pattern-recognition receptor signalling in health and disease. Nat. Rev. Immunol. 16, 35–50 (2016).

    Article  CAS  PubMed  Google Scholar 

  5. Zheng, Q. et al. Siglec1 suppresses antiviral innate immune response by inducing TBK1 degradation via the ubiquitin ligase TRIM27. Cell Res. 25, 1121–1136 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Portilho, D.M. et al. Endogenous TRIM5α function is regulated by SUMOylation and nuclear sequestration for efficient innate sensing in dendritic cells. Cell Rep. 14, 355–369 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Ansari, M.A. et al. Herpesvirus genome recognition induced acetylation of nuclear IFI16 is essential for its cytoplasmic translocation, inflammasome and IFN-b responses. PLoS Pathog. 11, e1005019 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Kim, T.W. et al. Transcriptional repression of IFN regulatory factor 7 by MYC is critical for type I IFN production in human plasmacytoid dendritic cells. J. Immunol. 197, 3348–3359 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Loo, Y.M. & Gale, M. Jr. Immune signaling by RIG-I-like receptors. Immunity 34, 680–692 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhang, Z. et al. The helicase DDX41 senses intracellular DNA mediated by the adaptor STING in dendritic cells. Nat. Immunol. 12, 959–965 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Parvatiyar, K. et al. The helicase DDX41 recognizes the bacterial secondary messengers cyclic di-GMP and cyclic di-AMP to activate a type I interferon immune response. Nat. Immunol. 13, 1155–1161 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, Z., Yuan, B., Lu, N., Facchinetti, V. & Liu, Y.J. DHX9 pairs with IPS-1 to sense double-stranded RNA in myeloid dendritic cells. J. Immunol. 187, 4501–4508 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Schröder, M., Baran, M. & Bowie, A.G. Viral targeting of DEAD box protein 3 reveals its role in TBK1/IKKepsilon-mediated IRF activation. EMBO J. 27, 2147–2157 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Linder, P. & Jankowsky, E. From unwinding to clamping - the DEAD box RNA helicase family. Nat. Rev. Mol. Cell Biol. 12, 505–516 (2011).

    Article  CAS  PubMed  Google Scholar 

  15. Calo, E. et al. RNA helicase DDX21 coordinates transcription and ribosomal RNA processing. Nature 518, 249–253 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Hoernes, T.P., Hüttenhofer, A. & Erlacher, M.D. mRNA modifications: dynamic regulators of gene expression? RNA Biol. 13, 760–765 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Wang, X. & He, C. Dynamic RNA modifications in posttranscriptional regulation. Mol. Cell 56, 5–12 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Desrosiers, R., Friderici, K. & Rottman, F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc. Natl. Acad. Sci. USA 71, 3971–3975 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. 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 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Liu, N. et al. N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature 518, 560–564 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  25. Zhao, X. et al. FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res. 24, 1403–1419 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

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

    Article  CAS  PubMed  Google Scholar 

  28. Cordin, O. & Beggs, J.D. RNA helicases in splicing. RNA Biol. 10, 83–95 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Liang, W.W. & Cheng, S.C. A novel mechanism for Prp5 function in prespliceosome formation and proofreading the branch site sequence. Genes Dev. 29, 81–93 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Bourgeois, C.F., Mortreux, F. & Auboeuf, D. The multiple functions of RNA helicases as drivers and regulators of gene expression. Nat. Rev. Mol. Cell Biol. 17, 426–438 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Putnam, A.A. & Jankowsky, E. DEAD-box helicases as integrators of RNA, nucleotide and protein binding. Biochim. Biophys. Acta 1829, 884–893 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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 

  33. Xu, Y.Z. et al. Prp5 bridges U1 and U2 snRNPs and enables stable U2 snRNP association with intron RNA. EMBO J. 23, 376–385 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Hirabayashi, R., Hozumi, S., Higashijima, S. & Kikuchi, Y. Ddx46 is required for multi-lineage differentiation of hematopoietic stem cells in zebrafish. Stem Cells Dev. 22, 2532–2542 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hozumi, S. et al. DEAD-box protein Ddx46 is required for the development of the digestive organs and brain in zebrafish. PLoS One 7, e33675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zarnack, K. et al. Direct competition between hnRNP C and U2AF65 protects the transcriptome from the exonization of Alu elements. Cell 152, 453–466 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Weber, M. et al. Incoming RNA virus nucleocapsids containing a 5′-triphosphorylated genome activate RIG-I and antiviral signaling. Cell Host Microbe 13, 336–346 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sun, X. et al. Nuclear retention of full-length HTT RNA is mediated by splicing factors MBNL1 and U2AF65. Sci. Rep. 5, 12521 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Chen, L.L. & Carmichael, G.G. Altered nuclear retention of mRNAs containing inverted repeats in human embryonic stem cells: functional role of a nuclear noncoding RNA. Mol. Cell 35, 467–478 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Boutz, P.L., Bhutkar, A. & Sharp, P.A. Detained introns are a novel, widespread class of post-transcriptionally spliced introns. Genes Dev. 29, 63–80 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Bahar Halpern, K. et al. Nuclear retention of mRNA in mammalian tissues. Cell Rep. 13, 2653–2662 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kufel, J., Bousquet-Antonelli, C., Beggs, J.D. & Tollervey, D. Nuclear pre-mRNA decapping and 5′ degradation in yeast require the Lsm2-8p complex. Mol. Cell. Biol. 24, 9646–9657 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, X. et al. Methyltransferase Dnmt3a upregulates HDAC9 to deacetylate the kinase TBK1 for activation of antiviral innate immunity. Nat. Immunol. 17, 806–815 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Wang, P. et al. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 344, 310–313 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Liu, J. et al. Rhbdd3 controls autoimmunity by suppressing the production of IL-6 by dendritic cells via K27-linked ubiquitination of the regulator NEMO. Nat. Immunol. 15, 612–622 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Huppertz, I. et al. iCLIP: protein-RNA interactions at nucleotide resolution. Methods 65, 274–287 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Moore, M.J. et al. Mapping Argonaute and conventional RNA-binding protein interactions with RNA at single-nucleotide resolution using HITS-CLIP and CIMS analysis. Nat. Protoc. 9, 263–293 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Peritz, T. et al. Immunoprecipitation of mRNA-protein complexes. Nat. Protoc. 1, 577–580 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Katz, Y., Wang, E.T., Airoldi, E.M. & Burge, C.B. Analysis and design of RNA sequencing experiments for identifying isoform regulation. Nat. Methods 7, 1009–1015 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Miller, J.C. et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 29, 143–148 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank T. Fang, M. Jin and Y. Li for technical assistance; X. Liu, C. Han, S. Xu, Y. Han and T. Chen for discussions; and the SIDANSAI Biotechnology Company (Shanghai, China) for helping to generate Ddx46+/− mice. Supported by the National Key Basic Research Program of China (2013CB530503 and 2012CB518900), the National Natural Science Foundation of China (31390431, 81422037 and 81671564) and the CAMS Innovation Fund for Medical Sciences (2016-12M-1-003).

Author information

Authors and Affiliations

Authors

Contributions

Q.Z., J.H., Y.Z., and Z.L. performed the experiments; Q.Z., J.H. and X.C. analyzed data and wrote the paper; and X.C. designed and supervised the research.

Corresponding author

Correspondence to Xuetao Cao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 DDX46 negatively regulates the production of type I interferons after viral infection.

(a) qRT-PCR analysis of Ifnb mRNA in macrophages transfected with the indicated siRNAs after infected with VSV (MOI=10) as indicated time. (b) Confocal microscopy of macrophages stained with DDX46 antibodies. Scale bar, 10 μm. (c,d) qRT-PCR and immunoblot analysis of DDX46 in macrophages transfected with NC, DDX46 siRNA#1 or #2 as indicated for 72 h. (e) qRT-PCR analysis of Ifnb mRNA expression in human THP1 cells transfected with NC or DDX46 siRNA for 72 h and infected with VSV (MOI=10) for indicated time. (f,g) Immunoblot analysis of the indicated protein in RAW264.7 cell clones stably overexpressing DDX46 (f) or transfected with siRNA for 72 h, and infected with VSV (MOI=10) for indicated time (g). (h,i) qRT-PCR analysis of Ifnb, Il6, Tnf and Il1b mRNA in RAW264.7 cells transfected with NC or DDX46 siRNA for 72 h and infected with VSV (MOI=10) (h) or treated with LPS (100ng/mL) (i) as indicated time. (j) Luciferase activity in HEK293T cells after co-transfection with DDX46 and the indicated plasmids. Luciferase activity is presented relative to Renilla luciferase activity. (k-m) Immunoblot analysis of DDX46 in mouse macrophages infected with VSV (k) or HSV (l) or in mouse peripheral blood mononuclear cells (PBMNC), myeloid conventional dendritic cells (cDC), plasmacytoid dendritic cells (pDC) and in human THP1 cells infected with VSV (m) as indicated time. NS: not significant; **p<0.01, *p<0.05 (Student’s t-test). Data are representative of three independent experiments (mean and s.d. of technical triplicates (a,c,e,h -j)) or from three independent experiments with similar results (b,d,f,g,k-m).

Supplementary Figure 2 Gene-ontology (GO) and KEGG analysis of DDX46-bound mRNA with the DAVID tool.

(a) Gel electrophoresis analysis of cDNA for removing free RT primers and size selection. (b) Gel image of amplified cDNA libraries and extraction of the indicated size ranges. (c,d) Overlap of two biological replicates for iCLIP in uninfected or infected RAW264.7 cells. Numbers show the sum of genes identified in each sample. (e) Overlap of transcripts identified by iCLIP of DDX46 in uninfected or infected RAW264.7 cells. Numbers in c,d,e show the sum of genes identified in each sample. (f,g) GO and KEGG analysis of the 1000 most enriched DDX46-bound mRNAs in uninfected cells but not in that of the infected cells. (h,i) GO and KEGG analysis of the 1000 most enriched DDX46-bound mRNAs in infected cell but not in that of uninfected cell. * indicates the enriched antiviral signaling pathway upon viral infection. Data are representative of three independent experiments with similar results (a,b).

Supplementary Figure 3 DDX46 associates with ALKBH5 to inhibit the expression of antiviral transcripts after viral infection.

(a) Fold enrichment of Gapdh and Ticam1 transcripts in DDX46-RNA co-immunoprecipitation versus RNA-protein input control in RAW264.7 infected with VSV (MOI=10). (b) The relative changes in PSI values observed in 1449 transcripts between wildtype and DDX46+/− macrophages by RNA-seq. (c) RAW264.7 cells were transfected with plasmids for 24 h, and then the cells were fixed, permeabilized, and stained with FISH probes for Tbk1 transcript. Scale bar, 10 μm. (d) RAW264.7 cells were fixed, permeabilized, and stained with FISH probes for Mavs transcript, and then stained with DDX46 and SC-35 antibody. Scale bar, 10 μm. (e) Immunoblot analysis of the indicated protein in macrophages transfected with NC or siRNA for 72 h, and infected with VSV (MOI=10) for indicated time. (f) Immunoblot analysis of the indicated protein in RAW264.7 cells transfected with NC or siRNA for 72 h, and infected with VSV (MOI=10) for indicated time. NS: not significant. (Student’s t-test). Data are representative of three independent experiments (mean and s.d. of technical triplicates (a)) or from three independent experiments with similar results (c-f).

Supplementary Figure 4 DDX46 associates with ALKBH5 to inhibit MAVS expression after viral infection.

(a) PAGE gel resolution of immunoprecipitated DDX46 and its associated proteins from peritoneal macrophages infected by VSV for the indicated time. Different bands were analyzed by MS. Arrow indicates the band of the protein detected by MS. (b) IP and immunoblot analysis of HEK293T cells after co-transfection of DDX46 with ALKBH5 upon various stimulation as indicated. (c) Luciferase activity (left) and immunoblot (right) analysis in HEK293T cells after co-transfection with MAVS, SRPK2, and ALKBH5 as indicated. (d) IP and immunoblot analysis of HEK293T cells after co-transfection of DDX46 mutants with ALKBH5 as indicated upon VSV (MOI=10) infection for 6 h. (e,f) Luciferase activity (left) and immunoblot (right) analysis in HEK293T cells after co-transfection with MAVS and DDX46 mutants or truncates as indicated. NS: not significant; *p<0.01 (Student’s t-test). Data are representative of three independent experiments (mean and s.d. of technical triplicates (c(left),e(left), f(left))) or from three independent experiments with similar results (a,b,c(right), d,e(right), f(right)).

Supplementary Figure 5 DDX46-inhibited production od type I interferons is dependent on the recruitment of ALKBH5.

(a,b) qRT-PCR analysis of Ifnb mRNA in RAW264.7 cells transfected with plasmids plus siRNAs as indicated, and infected with VSV (MOI=10) for 12 h. (c) Immunoblot analysis of ALKBH5 in Alkbh5−/− RAW264.7 cells. (d) Fold enrichment of the indicated mRNA transcripts in m6A in vitro IP versus mRNA input control. (e) RAW264.7 cells were fixed, permeabilized, and stained with FISH probes for MAVS transcript, and then stained with DDX46 and ALKBH5 antibody. Scale bar, 10 μm. NS: not significant; *p<0.01 (Student’s t-test). Data are representative of three independent experiments (mean and s.d. of technical triplicates (a,b,d)) or from three independent experiments with similar results (c, e).

Supplementary Figure 6 DDX46 negatively regulates the production of inflammatory cytokines and Ifnb in macrophage.

(a) qRT-PCR analysis of Il6, Tnf and Il1b mRNA in peritoneal macrophage cells from Ddx46+/+ or Ddx46+/− mice infected with VSV (MOI=10) as indicated time. (b) qRT-PCR analysis of Ifnb, Il6, Tnf and Il1b mRNA in peritoneal macrophage cells from Ddx46+/+ or Ddx46+/− mice and treated with LPS (100ng/mL) for indicated time. (c) Working model for the mechanism of RNA helicase DDX46 which recruits ALKBH5 to inhibit antiviral innate immunity by nuclear entrapping m6A demethylated antiviral transcripts via m6A modification. Ac: Acetylation. *p<0.05 (Student’s t-test). Data are representative of three independent experiments (mean and s.d. of technical triplicates (a,b)).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1–7

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, Q., Hou, J., Zhou, Y. et al. The RNA helicase DDX46 inhibits innate immunity by entrapping m6A-demethylated antiviral transcripts in the nucleus. Nat Immunol 18, 1094–1103 (2017). https://doi.org/10.1038/ni.3830

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3830

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