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:

Human nuclear Dicer restricts the deleterious accumulation of endogenous double-stranded RNA

Subjects

Abstract

Dicer is a central enzymatic player in RNA-interference pathways that acts to regulate gene expression in nearly all eukaryotes. Although the cytoplasmic function of Dicer is well documented in mammals, its nuclear function remains obscure. Here we show that Dicer is present in both the nucleus and cytoplasm, and its nuclear levels are tightly regulated. Dicer interacts with RNA polymerase II (Pol II) at actively transcribed gene loci. Loss of Dicer causes the appearance of endogenous double-stranded RNA (dsRNA), which in turn leads to induction of the interferon-response pathway and consequent cell death. Our results suggest that Pol II–associated Dicer restricts endogenous dsRNA formation from overlapping noncoding-RNA transcription units. Failure to do so has catastrophic effects on cell function.

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: Dicer localizes in both the nucleus and cytoplasm of HEK293 cells.
Figure 2: Dicer interaction with Pol II is dsRNA dependent, and Dicer-chromatin association is transcription dependent.
Figure 3: Dicer affects levels of nascent transcripts and TGS.
Figure 4: Loss of Dicer leads to accumulation of long dsRNA.
Figure 5: Loss of Dicer leads to the accumulation of dsRNA that colocalizes with chromatin-associated Dicer loci.
Figure 6: Loss of Dicer leads to a reduction of siRNAs that colocalize with chromatin-associated Dicer sites and dsRNA sites.
Figure 7: Loss of Dicer triggers the interferon response.
Figure 8: Model of nuclear Dicer function.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Grewal, S.I. RNAi-dependent formation of heterochromatin and its diverse functions. Curr. Opin. Genet. Dev. 20, 134–141 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Bartel, D.P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Baulcombe, D. RNA silencing in plants. Nature 431, 356–363 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Gullerova, M. & Proudfoot, N.J. Cohesin complex promotes transcriptional termination between convergent genes in S. pombe. Cell 132, 983–995 (2008).

    Article  CAS  PubMed  Google Scholar 

  5. Gullerova, M. & Proudfoot, N.J. Convergent transcription induces transcriptional gene silencing in fission yeast and mammalian cells. Nat. Struct. Mol. Biol. 19, 1193–1201 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Minchiotti, G. & Di Nocera, P.P. Convergent transcription initiates from oppositely oriented promoters within the 5′ end regions of Drosophila melanogaster F elements. Mol. Cell. Biol. 11, 5171–5180 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Faghihi, M.A. & Wahlestedt, C. Regulatory roles of natural antisense transcripts. Nat. Rev. Mol. Cell Biol. 10, 637–643 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Samuel, C.E. Antiviral actions of interferons. Clin. Microbiol. Rev. 14, 778–809 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. García, M.A., Meurs, E.F. & Esteban, M. The dsRNA protein kinase PKR: virus and cell control. Biochimie 89, 799–811 (2007).

    Article  PubMed  Google Scholar 

  10. Han, J. et al. Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex. Cell 125, 887–901 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Yates, L.A., Norbury, C.J. & Gilbert, R.J. The long and short of microRNA. Cell 153, 516–519 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Jaskiewicz, L. & Filipowicz, W. Role of Dicer in posttranscriptional RNA silencing. Curr. Top. Microbiol. Immunol. 320, 77–97 (2008).

    CAS  PubMed  Google Scholar 

  13. Bühler, M., Verdel, A. & Moazed, D. Tethering RITS to a nascent transcript initiates RNAi- and heterochromatin-dependent gene silencing. Cell 125, 873–886 (2006).

    Article  PubMed  Google Scholar 

  14. Bühler, M., Haas, W., Gygi, S.P. & Moazed, D. RNAi-dependent and -independent RNA turnover mechanisms contribute to heterochromatic gene silencing. Cell 129, 707–721 (2007).

    Article  PubMed  Google Scholar 

  15. Moazed, D. et al. Studies on the mechanism of RNAi-dependent heterochromatin assembly. Cold Spring Harb. Symp. Quant. Biol. 71, 461–471 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Billy, E., Brondani, V., Zhang, H., Muller, U. & Filipowicz, W. Specific interference with gene expression induced by long, double-stranded RNA in mouse embryonal teratocarcinoma cell lines. Proc. Natl. Acad. Sci. USA 98, 14428–14433 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Provost, P. et al. Ribonuclease activity and RNA binding of recombinant human Dicer. EMBO J. 21, 5864–5874 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kotaja, N. et al. The chromatoid body of male germ cells: similarity with processing bodies and presence of Dicer and microRNA pathway components. Proc. Natl. Acad. Sci. USA 103, 2647–2652 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jakymiw, A. et al. Overexpression of dicer as a result of reduced let-7 microRNA levels contributes to increased cell proliferation of oral cancer cells. Genes Chromosom. Cancer 49, 549–559 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Doyle, M. et al. The double-stranded RNA binding domain of human Dicer functions as a nuclear localization signal. RNA 19, 1238–1252 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Haussecker, D. & Proudfoot, N.J. Dicer-dependent turnover of intergenic transcripts from the human β-globin gene cluster. Mol. Cell. Biol. 25, 9724–9733 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Morris, K.V., Chan, S.W., Jacobsen, S.E. & Looney, D.J. Small interfering RNA-induced transcriptional gene silencing in human cells. Science 305, 1289–1292 (2004).

    Article  CAS  PubMed  Google Scholar 

  23. Janowski, B.A. et al. Inhibiting gene expression at transcription start sites in chromosomal DNA with antigene RNAs. Nat. Chem. Biol. 1, 216–222 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Saint-André, V., Batsche, E., Rachez, C. & Muchardt, C. Histone H3 lysine 9 trimethylation and HP1γ favor inclusion of alternative exons. Nat. Struct. Mol. Biol. 18, 337–344 (2011).

    Article  PubMed  Google Scholar 

  25. Alló, M. et al. Control of alternative splicing through siRNA-mediated transcriptional gene silencing. Nat. Struct. Mol. Biol. 16, 717–724 (2009).

    Article  PubMed  Google Scholar 

  26. Ameyar-Zazoua, M. et al. Argonaute proteins couple chromatin silencing to alternative splicing. Nat. Struct. Mol. Biol. 19, 998–1004 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Schmitter, D. et al. Effects of Dicer and Argonaute down-regulation on mRNA levels in human HEK293 cells. Nucleic Acids Res. 34, 4801–4815 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nesterova, T.B. et al. Dicer regulates Xist promoter methylation in ES cells indirectly through transcriptional control of Dnmt3a. Epigenetics Chromatin 1, 2 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Morris, D.P., Michelotti, G.A. & Schwinn, D.A. Evidence that phosphorylation of the RNA polymerase II carboxyl-terminal repeats is similar in yeast and humans. J. Biol. Chem. 280, 31368–31377 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Zamore, P.D. & Haley, B. Ribo-gnome: the big world of small RNAs. Science 309, 1519–1524 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Kim, D.H., Villeneuve, L.M., Morris, K.V. & Rossi, J.J. Argonaute-1 directs siRNA-mediated transcriptional gene silencing in human cells. Nat. Struct. Mol. Biol. 13, 793–797 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Affymetrix ENCODE Transcriptome Project; Cold Spring Harbor Laboratory ENCODE Transcriptome Project. Post-transcriptional processing generates a diversity of 5′-modified long and short RNAs. Nature 457, 1028–1032 (2009).

  33. Wang, R. et al. Mouse embryonic stem cells are deficient in type I interferon expression in response to viral infections and double-stranded RNA. J. Biol. Chem. 288, 15926–15936 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stetson, D.B. & Medzhitov, R. Type I interferons in host defense. Immunity 25, 373–381 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Kawai, T. & Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34, 637–650 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Wilson, R.C. & Doudna, J.A. Molecular mechanisms of RNA interference. Annu. Rev. Biophys. 42, 217–239 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gagnon, K.T., Li, L., Chu, Y., Janowski, B.A. & Corey, D.R. RNAi factors are present and active in human cell nuclei. Cell Reports 6, 211–221 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Lee, J.T. Epigenetic regulation by long noncoding RNAs. Science 338, 1435–1439 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Kaneko, H. et al. DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature 471, 325–330 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Passon, N. et al. Expression of Dicer and Drosha in triple-negative breast cancer. J. Clin. Pathol. 65, 320–326 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Castel, S.E. & Martienssen, R.A. RNA interference in the nucleus: roles for small RNAs in transcription, epigenetics and beyond. Nat. Rev. Genet. 14, 100–112 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Peters, A.H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001).

    Article  CAS  PubMed  Google Scholar 

  43. Fodor, B.D. et al. Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev. 20, 1557–1562 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bulut-Karslioglu, A. et al. A transcription factor–based mechanism for mouse heterochromatin formation. Nat. Struct. Mol. Biol. 19, 1023–1030 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Martens, J.H. et al. The profile of repeat-associated histone lysine methylation states in the mouse epigenome. EMBO J. 24, 800–812 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shinkai, Y. & Tachibana, M. H3K9 methyltransferase G9a and the related molecule GLP. Genes Dev. 25, 781–788 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fagegaltier, D. et al. The endogenous siRNA pathway is involved in heterochromatin formation in Drosophila. Proc. Natl. Acad. Sci. USA 106, 21258–21263 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Aagaard, L. et al. A facile lentiviral vector system for expression of doxycycline-inducible shRNAs: knockdown of the pre-miRNA processing enzyme Drosha. Mol. Ther. 15, 938–945 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. Landt, S.G. et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Pall, G.S., Codony-Servat, C., Byrne, J., Ritchie, L. & Hamilton, A. Carbodiimide-mediated cross-linking of RNA to nylon membranes improves the detection of siRNA, miRNA and piRNA by northern blot. Nucleic Acids Res. 35, e60 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to E. Johnson for help with transmission electron microscopy and scanning electron microscopy experiments. This work was supported by grants from the Wellcome Trust (091805/Z/10/Z to N.J.P.) and E.P. Abraham Trust (to N.J.P.) and by a Medical Research Council Career Development Award (MR/K006606/1 to M.G.) and a L'Oreal-UNESCO Woman in Science UK and Ireland award (to M.G.).

Author information

Authors and Affiliations

Authors

Contributions

E.W. and M.G. performed all the experimental analyses. M.S. and K.K.-G. performed bioinformatics analyses of the ChIP-seq and RNA-seq data. E.W., M.G. and N.J.P. designed the experiments and wrote the manuscript.

Corresponding authors

Correspondence to Nick J Proudfoot or Monika Gullerova.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Dicer localizes in both nucleus and cytoplasm of HEK293 cells.

a) Western blot analysis of protein extracts from HEK293 cells with inducible integrated shRNA transgene. Induction by doxycycline leads to Dicer-specific shRNA production and consequent Dicer knockdown. Antibodies specific to Dicer and γ-actin were employed. b) Western blot analysis of Dicer in nuclear and cytoplasmic extracts from HEK293 cells transfected with episomal GFP-Dicer or RFP-Dicer expressing plasmids. Grp75 and γ-actin were used as controls. c) Expression of GFP-Dicer transgene (green) in normal and Dicer knockout murine embryonic stem cells as in Figure 1b. d) Control FRAP experiment shows Dicer-GFP (green) recovery in nucleus of HEK293 cells (lacking GFP signal) after bleaching. This provides a background control to FRAP analysis of GFP-Dicer expression in the nucleus of Dicer knockdown cells. Here, only background signals can be detected. The cell is shown in the left hand panel before bleaching. In the middle panel, the red circle corresponds to bleached area in nucleus. Fluorescent recovery was measured in the green circle. Fluorescent background values were measured in the blue circle. Fluorescent recovery was measured in 20 frames, bleaching was initiated at frame 5. Right-hand panel shows the cell after recovery. Absolute levels of fluorescence and relative levels of recovery (background fluorescence subtracted from fluorescence levels in measured region) are shown in the left and right panels, respectively. All experiments described in Supplementary Fig. 1a-c, were replicated three times.

Supplementary Figure 2 Genomic annotation of Dicer ChIP-seq peaks.

a) Table showing the top 12 peaks of Dicer binding loci according to the ChIP sequencing data. Nearest gene identity is shown. b) The 4 additional loci chosen for further analysis, showing a range of Dicer binding c) Metagene analysis of top 118 Dicer binding sites (ChIP-seq peaks) showing their location distribution around TSS and PAS of coding genes in the human genome. Data were grouped into 250 nt bins. Each bin-midpoint was plotted on the graph. d) Table summarising the enrichment of repetitive DNA sequence elements in the Dicer ChIP-seq data. The RepeatMasker hg19 table was used to extract genomic repetitive elements. Repetitive element overlapping a Dicer peak with 5 nt or more was included in the analysis. Each category of repetitive elements as presented, was analysed separately. p-values are based on a background distribution of sampling 118 random genomic sequences of the same length as Dicer peaks. Sampling was performed more than 1000 times. e) H3 control ChIP experiment as in Figure 2e. f) Top panel: Diagram of the MTRNR2L6. The start site of transcription (arrow) and the position of the PCR amplicons (bold) are indicated. Bottom panels: Pol II, Dicer and H3 ChIP experiments as in Figure 2e.

Supplementary Figure 3 Dicer and Pol II binding affect each other at selected loci.

a) qRT-PCR as in Fig 3d and 3e. Probes were specific to GAPDH and b) 28S rDNA.

Supplementary Figure 4 Loss of Drosha has no effect on the accumulation of long dsRNA.

a) IF as in Fig. 1a using anti-Drosha and J2 antibodies on normal and Drosha knockdown HEK293 cells. b) Levels of pri-mRNA15a transcripts as determined by qRT-PCR analysis using a specific primer for reverse transcription. RNA was isolated from normal HEK293 cells and cells treated with an shRNA against Drosha for 10 days. Signals are based on average values ± s.d. from three independent biological experiments and are normalised to the levels of transcripts in normal cells, set as 1.

Supplementary Figure 5 Loss of Dicer leads to accumulation of long dsRNA.

IF as in Fig. 1a using anti-Dicer and J2 antibodies on Dicer knockout murine embryonic stem cells.

Supplementary Figure 6 Loss of Dicer leads to accumulation of dsRNA.

a) Histogram of peak summit ratio of dsRNA in Dicer knockdown cells to dsRNA in normal cells. Any dsRNA signal >10 in both uninduced and induced cells was considered. Value higher than 1 indicates accumulation of dsRNA and value lower than 1 indicates a decrease in dsRNA levels in Dicer knockdown cells. The black line marks the value of 1. Dark blue bins indicate the number of times a ratio falls in value span marked on the horizontal axis. Light blue bins represent a cumulative number for the values under 1 or over 1 separately. Centromeric repeat regions were excluded from analysis. b) Diagram of the change level in dsRNA (according to peak summit value) accumulation in normal and Dicer knockdown cells. Any dsRNA signal >10 in both uninduced and induced cells was considered. Plotted levels from normal cells are subtracted Dicer knockdown cell levels. Value less than 0 (marked by a black vertical line) indicates the absolute value of the decrease in dsRNA, while a value greater than 0 indicates an increase. Each blue point depicts one locus of Dicer-dsRNA co-localization. The loci were ordered according to the difference value in ascending order. One point is too large to be depicted in the plot, it has value 2432530. Note the scale of the graph is (x104). Centromeric repeat regions were excluded from analysis. c) Box Plot of change in dsRNA levels (according to peak summit value) between Dicer knockdown and normal cells. Any dsRNA signal >10 in both uninduced and induced cells was considered. The red mark indicated the median, at a value of 855. The lower quartile q1 (25th percentile) is at -19 and the upper quartile q3 (75th percentile) at 3992 (blue box). The shift in the positive direction of the box shows a general increase in dsRNA at Dicer loci. 20 outlier values were excluded from the plot. Centromeric repeat regions were excluded from analysis. d) Tabular summary of enrichment of repetitive elements in dsRNA-seq data. The RepeatMasker hg19 table was used to extract genomic repetitive elements. Repetitive element overlapping a dsRNA peak with 5 nt or more was included in the analysis. Each category of repetitive elements as presented, was analysed separately. p-values are based on a background distribution of sampling 118 random genomic sequences of the same length as Dicer peaks. Sampling was performed more than 1000 times. e) Metagene analysis of top dsRNA peaks (see Online Methods) showing location distribution around TSS and PAS of coding genes in the human genome. Data were grouped into 250 nt bins. Each bin-midpoint was plotted on graph.

Supplementary Figure 7 Loss of Dicer triggers the interferon response.

a) Quantitation of Western blot analysis of cell extracts isolated from normal and Dicer knockdown HEK293 cells, growing in tissue culture for 1 or 2 weeks using antibodies specific to Dicer, TLR3, PKR1 and γ-actin (see Figure 7a). Signals were measured by image-Quant software, are expressed as a % of input and are expressed as an average of three independent experiments, normalised to normal HEK293 cell levels, set as 1. * indicates statistical significance (p < 0.05), based on unpaired, two-tailed distribution Student's t-test. b) IF as in Figure 4c using anti-PKR antibody (green).

Supplementary Figure 8 Loss of Dicer triggers the interferon response.

a) Morphological changes of normal and Dicer knockdown HEK293 cells using Electron Microscopy (SEM) at 3000-fold magnification. b) TEM analysis of nuclear structure in normal and Dicer knockdown HEK293. c) TEM as in middle panel focusing on mitochondria. Representative images from 20 cells are presented.

Supplementary Figure 9 Uncropped blot images.

a) Relating to Figure 2. b) Relating to Figure 4. c) Relating to Figure 6. d) Relating to Figure 7.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Table 1 (PDF 6583 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

White, E., Schlackow, M., Kamieniarz-Gdula, K. et al. Human nuclear Dicer restricts the deleterious accumulation of endogenous double-stranded RNA. Nat Struct Mol Biol 21, 552–559 (2014). https://doi.org/10.1038/nsmb.2827

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2827

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