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:

RNA helicase DDX21 coordinates transcription and ribosomal RNA processing

Abstract

DEAD-box RNA helicases are vital for the regulation of various aspects of the RNA life cycle1, but the molecular underpinnings of their involvement, particularly in mammalian cells, remain poorly understood. Here we show that the DEAD-box RNA helicase DDX21 can sense the transcriptional status of both RNA polymerase (Pol) I and II to control multiple steps of ribosome biogenesis in human cells. We demonstrate that DDX21 widely associates with Pol I- and Pol II-transcribed genes and with diverse species of RNA, most prominently with non-coding RNAs involved in the formation of ribonucleoprotein complexes, including ribosomal RNA, small nucleolar RNAs (snoRNAs) and 7SK RNA. Although broad, these molecular interactions, both at the chromatin and RNA level, exhibit remarkable specificity for the regulation of ribosomal genes. In the nucleolus, DDX21 occupies the transcribed rDNA locus, directly contacts both rRNA and snoRNAs, and promotes rRNA transcription, processing and modification. In the nucleoplasm, DDX21 binds 7SK RNA and, as a component of the 7SK small nuclear ribonucleoprotein (snRNP) complex, is recruited to the promoters of Pol II-transcribed genes encoding ribosomal proteins and snoRNAs. Promoter-bound DDX21 facilitates the release of the positive transcription elongation factor b (P-TEFb) from the 7SK snRNP in a manner that is dependent on its helicase activity, thereby promoting transcription of its target genes. Our results uncover the multifaceted role of DDX21 in multiple steps of ribosome biogenesis, and provide evidence implicating a mammalian RNA helicase in RNA modification and Pol II elongation control.

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: DDX21 associates with actively transcribed ribosomal genes.
Figure 2: DDX21 chromatin association is sensitive to Pol I and Pol II transcriptional status.
Figure 3: DDX21 binds rRNA and snoRNAs and facilitates rRNA modification.
Figure 4: DDX21 promotes release of P-TEFb from the 7SK snRNP.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

All sequencing data have been deposited in Gene Expression Omnibus (GEO) data repository under accession number GSE56802.

References

  1. Rocak, S. & Linder, P. DEAD-box proteins: the driving forces behind RNA metabolism. Nature Rev. Mol. Cell Biol. 5, 232–241 (2004)

    Article  CAS  Google Scholar 

  2. Russell, R., Jarmoskaite, I. & Lambowitz, A. M. Toward a molecular understanding of RNA remodeling by DEAD-box proteins. RNA Biol. 10, 44–55 (2013)

    Article  CAS  Google Scholar 

  3. 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  Google Scholar 

  4. Henning, D., So, R. B., Jin, R., Lau, L. F. & Valdez, B. C. Silencing of RNA helicase II/Guα inhibits mammalian ribosomal RNA production. J. Biol. Chem. 278, 52307–52314 (2003)

    Article  CAS  Google Scholar 

  5. Yang, H. et al. Down-regulation of RNA helicase II/Gu results in the depletion of 18 and 28 S rRNAs in Xenopus oocyte. J. Biol. Chem. 278, 38847–38859 (2003)

    Article  CAS  Google Scholar 

  6. Westermarck, J. et al. The DEXD/H-box RNA helicase RHII/Gu is a co-factor for c-Jun-activated transcription. EMBO J. 21, 451–460 (2002)

    Article  CAS  Google Scholar 

  7. Zentner, G. E., Saiakhova, A., Manaenkov, P., Adams, M. D. & Scacheri, P. C. Integrative genomic analysis of human ribosomal DNA. Nucleic Acids Res. 39, 4949–4960 (2011)

    Article  CAS  Google Scholar 

  8. Cong, R. et al. Interaction of nucleolin with ribosomal RNA genes and its role in RNA polymerase I transcription. Nucleic Acids Res. 40, 9441–9454 (2012)

    Article  Google Scholar 

  9. Dieci, G., Preti, M. & Montanini, B. Eukaryotic snoRNAs: a paradigm for gene expression flexibility. Genomics 94, 83–88 (2009)

    Article  CAS  Google Scholar 

  10. Chao, S. H. et al. Flavopiridol inhibits P-TEFb and blocks HIV-1 replication. J. Biol. Chem. 275, 28345–28348 (2000)

    Article  CAS  Google Scholar 

  11. Chao, S. H. & Price, D. H. Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. J. Biol. Chem. 276, 31793–31799 (2001)

    Article  CAS  Google Scholar 

  12. Valdez, B. C., Henning, D., Perumal, K. & Busch, H. RNA-unwinding and RNA-folding activities of RNA helicase II/Gu–two activities in separate domains of the same protein. Eur. J. Biochem. 250, 800–807 (1997)

    Article  CAS  Google Scholar 

  13. Perlaky, L., Valdez, B. C. & Busch, H. Effects of cytotoxic drugs on translocation of nucleolar RNA helicase RH-II/Gu. Exp. Cell Res. 235, 413–420 (1997)

    Article  CAS  Google Scholar 

  14. Drygin, D. et al. Anticancer activity of CX-3543: a direct inhibitor of rRNA biogenesis. Cancer Res. 69, 7653–7661 (2009)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. 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  Google Scholar 

  17. Lui, L. & Lowe, T. Small nucleolar RNAs and RNA-guided post-transcriptional modification. Essays Biochem. 54, 53–77 (2013)

    Article  CAS  Google Scholar 

  18. Hughes, J. M. & Ares, M., Jr Depletion of U3 small nucleolar RNA inhibits cleavage in the 5′ external transcribed spacer of yeast pre-ribosomal RNA and impairs formation of 18S ribosomal RNA. EMBO J. 10, 4231–4239 (1991)

    Article  CAS  Google Scholar 

  19. Fatica, A., Galardi, S., Altieri, F. & Bozzoni, I. Fibrillarin binds directly and specifically to U16 box C/D snoRNA. RNA 6, 88–95 (2000)

    Article  CAS  Google Scholar 

  20. Newman, D. R., Kuhn, J. F., Shanab, G. M. & Maxwell, E. S. Box C/D snoRNA-associated proteins: two pairs of evolutionarily ancient proteins and possible links to replication and transcription. RNA 6, 861–879 (2000)

    Article  CAS  Google Scholar 

  21. Petfalski, E., Dandekar, T., Henry, Y. & Tollervey, D. Processing of the precursors to small nucleolar RNAs and rRNAs requires common components. Mol. Cell. Biol. 18, 1181–1189 (1998)

    Article  CAS  Google Scholar 

  22. Yu, Y. T., Shu, M. D. & Steitz, J. A. A new method for detecting sites of 2′-O-methylation in RNA molecules. RNA 3, 324–331 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang, Z., Zhu, Q., Luo, K. & Zhou, Q. The 7SK small nuclear RNA inhibits the CDK9/cyclin T1 kinase to control transcription. Nature 414, 317–322 (2001)

    Article  ADS  CAS  Google Scholar 

  24. Ji, X. et al. SR proteins collaborate with 7SK and promoter-associated nascent RNA to release paused polymerase. Cell 153, 855–868 (2013)

    Article  CAS  Google Scholar 

  25. McNamara, R. P., McCann, J. L., Gudipaty, S. A. & D’Orso, I. Transcription factors mediate the enzymatic disassembly of promoter-bound 7SK snRNP to locally recruit P-TEFb for transcription elongation. Cell Rep. 5, 1256–1268 (2013)

    Article  CAS  Google Scholar 

  26. Castelo-Branco, G. et al. The non-coding snRNA 7SK controls transcriptional termination, poising, and bidirectionality in embryonic stem cells. Genome Biol. 14, R98 (2013)

    Article  Google Scholar 

  27. Nguyen, V. T., Kiss, T., Michels, A. A. & Bensaude, O. 7SK small nuclear RNA binds to and inhibits the activity of CDK9/cyclin T complexes. Nature 414, 322–325 (2001)

    Article  ADS  CAS  Google Scholar 

  28. Peterlin, B. M. & Price, D. H. Controlling the elongation phase of transcription with P-TEFb. Mol. Cell 23, 297–305 (2006)

    Article  CAS  Google Scholar 

  29. Michels, A. A. et al. Binding of the 7SK snRNA turns the HEXIM1 protein into a P-TEFb (CDK9/cyclin T) inhibitor. EMBO J. 23, 2608–2619 (2004)

    Article  CAS  Google Scholar 

  30. Liu, W. et al. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 155, 1581–1595 (2013)

    Article  CAS  Google Scholar 

  31. Flynn, R. A., Almada, A. E., Zamudio, J. R. & Sharp, P. A. Antisense RNA polymerase II divergent transcripts are P-TEFb dependent and substrates for the RNA exosome. Proc. Natl Acad. Sci. USA 108, 10460–10465 (2011)

    Article  ADS  CAS  Google Scholar 

  32. Wassarman, D. A. & Steitz, J. A. Structural analyses of the 7SK ribonucleoprotein (RNP), the most abundant human small RNP of unknown function. Mol. Cell. Biol. 11, 3432–3445 (1991)

    Article  CAS  Google Scholar 

  33. Sharma, A. et al. The Werner syndrome helicase is a cofactor for HIV-1 long terminal repeat transactivation and retroviral replication. J. Biol. Chem. 282, 12048–12057 (2007)

    Article  CAS  Google Scholar 

  34. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nature Biotechnol. 28, 495–501 (2010)

    Article  CAS  Google Scholar 

  35. Carey, M. F., Peterson, C. L. & Smale, S. T. Dignam and Roeder nuclear extract preparation. Cold Spring Harb. Protoc. 2009 10.1101/pdb.prot5330 (2009)

  36. Konig, J. et al. iCLIP–transcriptome-wide mapping of protein–RNA interactions with individual nucleotide resolution. J. Vis. Exp. 50, 2638 (2011)

    Google Scholar 

  37. Krueger, B. J., Varzavand, K., Cooper, J. J. & Price, D. H. The mechanism of release of P-TEFb and HEXIM1 from the 7SK snRNP by viral and cellular activators includes a conformational change in 7SK. PLoS ONE 5, e12335 (2010)

    Article  ADS  Google Scholar 

Download references

Acknowledgements

We thank D. H. Price for the LARP7 antibody, K. Lane from M. Covert’s laboratory for metabolic inhibitors, K. Cimprich and members of the Chang and Wysocka laboratories for discussions, and B. Zarnegar and P. Khavari for discussions regarding iCLIP. This work was supported by the Stanford Medical Scientist Training Program and T32CA09302 (R.A.F.), AP Giannini Foundation (R.C.S.), National Institutes of Health grants R01-HG004361, R01-ES023168, P50-HG007735 (H.Y.C.) and R01-GM095555 (J.W.), W. M. Keck Foundation (J.W.), and Helen Hay Whitney Foundation (E.C.). H.Y.C. is an Early Career Scientist of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

H.Y.C. and J.W. supervised the project; E.C. and R.A.F. conceived and designed the study; E.C. and R.A.F. performed experiments and analysed ChIP-seq data. R.A.F. performed iCLIP and L.M., R.C.S. and R.A.F. analysed iCLIP data; R.A.F., E.C., J.W. and H.Y.C. wrote the manuscript with input from all co-authors.

Corresponding authors

Correspondence to Howard Y. Chang or Joanna Wysocka.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 DDX21 associates with non- and protein-coding ribosomal genes.

a, MEME analysis of DDX21-bound regions defined by DDX21 ChIP-seq. Motif logo, annotated transcription factor, number of motif instances within the ChIP-seq regions, Z score, and P value for each motif are shown. b, DDX21 ChIP-qPCR from HeLa cell chromatin extracts with primers spanning a representative number of loci found to be enriched in the DDX21 ChIP-seq analyses from HEK293 cells. Data are mean and s.d. of three independent experiments. c, Comparison of DDX21 (this study) and H3K4me3 (publically available data, see Methods for accession numbers) ChIP-seq-bound regions. 2,863 regions are common between the data sets, 505 regions are unique to DDX21, and 11,403 regions are unique to the H3K4me3 data set. d, Gene Ontology terms for H3K4me3 regions that are either DDX21-bound (left) or not bound by DDX21 (right). e, Box plots representing the expression levels of snoRNA-host genes whose promoter regions are either bound or not by DDX21. As shown, snoRNA-host gene promoters bound by DDX21 are, on average, more highly expressed than those not occupied by DDX21. Fragments per kilobase of exon per million mapped reads (FPKM) values were taken from publically available HEK293 RNA-seq data (see Methods for accession number). The P value (P ≤ 0.05) was calculated using the Wilcoxon signed-rank test. f, g, UCSC genome browser tracks depicting DDX21 ChIP-seq and iCLIP-seq, and RNA-seq enrichment profiles at differentially expressed snoRNA-host genes in HEK293 cells.

Extended Data Figure 2 DDX21 positively regulates transcription of Pol I- and Pol II-dependent ribosomal genes.

a, siRNA-mediated knockdown of the DDX21 antibody used for ChIP. We transfected HEK293 cells with two different sets of siRNAs targeting endogenous DDX21 mRNA (siRNA1 and siRNA2 (3′ UTR)) and performed western blots with the indicated antibodies. As shown, the DDX21-specific band is diminished in cells transfected with DDX21-targeting siRNAs, but not with control siRNAs. Actin was used as a loading control for this experiment. b, RT–qPCR analysis assessing the RNA expression levels of the same genes analysed in Fig. 1h upon DDX21 knockdown by a second siRNA that targets the 3′ UTR of DDX21 mRNA. Data are mean and s.d. of three independent experiments. For DDX21-target genes the difference between control and DDX21 siRNA is significant, P  0.05 (Student’s t-test). c, Diagram of DDX21 protein domains. The two conserved RecA-like (A and B) domains and the GUCT domains are shown in green and blue, respectively. Amino acids targeted for mutation12 to convert DDX21WT into DDX21SAT, the ATP-hydrolysis mutant, are indicated with red and purple lines in the diagram. Specific amino acid changes are displayed below. d, qRT–PCR analysis assessing nascent unspliced mRNA levels from additional DDX21-target and DDX21-non-target promoters. For a detailed description see Fig. 1j. Data are mean and s.d. of three biological replicates. e, Nuclear rRNA abundance analysis by RNA BioAnalyzer of HEK293 cells depleted of DDX21 and rescued with DDX21WT, DDX21SAT, or DDX21DEV. For each analysis, total RNA was isolated from 1,500,000 nuclei. Total nanogram amounts are shown for each of the two large rRNA subunits.

Extended Data Figure 3 Selective inhibition of Pol I alters DDX21 nuclear localization and chromatin association.

a, b, Immunofluorescence images of methanol-fixed HEK293 cells after 1 h incubation with either DMSO or 2 μM of the specific Pol I inhibitor CX-5461. DDX21 (a) and fibrillarin (b) immuno-labellings are shown. Scale bars, 10 μm. c, d, ChIP-qPCR analyses from HEK293 sampling DDX21 genomic occupancy, at the rDNA locus (c) and at a representative panel of Pol II-regulated gene promoters (d), after treatment with DMSO or CX-5461. Data are mean and s.d. of three independent experiments. As displayed, inhibition of Pol I alters DDX21 nuclear localization and this coincides with nearly complete eviction of DDX21 from Pol I- and Pol II-regulated genes. e, f, ChIP-qPCR analyses from HEK293 cells treated with 50 ng ml−1 of actinomycin-D for 1 h. Binding of the transcriptional repressor CTCF across the rDNA locus (e) and the c-MYC insulator element (MINE) (f) demonstrates that actinomycin-D treatment does not effect CTCF binding to chromatin. Red arrow indicates relative location of the CTCF DNA-binding site (DBS) at the rDNA locus. Data are mean and s.d. of three independent experiments.

Extended Data Figure 4 DDX21 nuclear re-localization is preferentially sensitive to acute transcriptional inhibition over other cellular stressors.

Immunofluorescence analyses of HEK293 cells after targeting different metabolic pathways. For inhibition of mitogen, cells were starved for 16 h in the absence of serum. For cellular respiration inhibition, cells were treated for with either oligomycin (100 μM) or 2-deoxy-d-glucose (10 mM) for 1 h. To inhibit the mTOR pathway, cells were treated with 250 nM of either Torin 1 or rapamycin for 2 h.

Extended Data Figure 5 Tandem affinity iCLIP of FH–DDX21WT.

a, FH–DDX21WT iCLIP 32P-autoradiogram and western blots. All samples were loaded with constant input lysate amounts (actin loading). FH–DDX21WT was isolated from HEK293 cells induced to express the transgene for 24 h and crosslinked with ultraviolet light (top panel same as Fig. 3a). b, Schematic of the modified iCLIP procedure. To achieve high stringency and specificity Flag–HA–DDX21WT is first purified on anti-Flag–M2 agarose beads, washed with 1 M NaCl, 1% Triton X-100 and 1% sodium deoxycholate. Complexes are specifically eluted with Flag peptide and recaptured with anti-HA agarose. Standard iCLIP steps were performed thereafter to generate deep sequencing libraries. ce, Scatter plot analysis of iCLIP reverse transcription stops on snoRNAs, rRNA and mRNAs within the FH–DDX21WT (this study) and hnRNP-C (ref. 16; publically available data) data sets. Little concordance between the data sets is evident, suggesting specific transcriptome targets of these two RNA binding proteins (RBPs). f, DDX21WT ultraviolet RNA immunoprecipitation qRT–PCR of FH–DDX21WT was performed in three conditions: native HEK293 cells crosslinked with ultraviolet light; FH–DDX21WT HEK293 cells without crosslinking; and FH–DDX21WT HEK293 cells with ultraviolet crosslinking. snoRNAs, scaRNAs and TERC were validated targets identified in the sequencing data. Each experiment was performed in biological duplicates (rep1 and rep2) and error bars represent s.d. of technical triplicates.

Extended Data Figure 6 Tandem affinity iCLIP of FH–DDX21SAT.

a, FH–DDX21SAT was isolated from HEK293 cells induced to express the transgene for 24 h, at which point we did not observe significant dominant negative effects. iCLIP was performed as described for FH–DDX21WT, and biological duplicates of FH–DDX21SAT iCLIP 32P-autoradiogram and western blots (lanes 2 and 3) are shown. All samples were loaded with constant input lysate amounts (actin loading). FH–DDX21WT was loaded as a control. WB, western blot. b, Left, DDX21SAT iCLIP reads annotated to known repetitive (rRNA and snRNAs) and non-repetitive (hg19 genome build: mRNAs and snoRNAs) regions of the human genome. Categories are notes with their respective percentage of the total iCLIP experiment. Right, enriched Gene Ontology and KEGG pathway terms from DDX21SAT-bound mRNAs obtained using the DAVID tool. The x axis values (in log scale) correspond to the negative Benjamini P value. c, Distribution of all DDX21SAT-bound snoRNAs, representing C/D box, H/ACA box and scaRNAs. The number (n) and fraction (per cent) of each snoRNA type is displayed. d, Comparison of the snoRNAs bound by DDX21WT and DDX21SAT, revealing significant overlap between the active and catalytically inactive DDX21. e, DDX21SAT iCLIP reads mapped to the transcribed region of the rDNA. f, DDX21WT (left) and DDX21SAT (right) iCLIP reads mapped to the repetitive U3 snoRNA. Binding is represented as reverse transcription stops per nucleotide normalized to the total number of reverse transcription stops mapping to the U3 snoRNA. Two strong binding sites are evident between bases 25–40 and 175–185 of U3 in DDX21WT iCLIP, whereas the 5′ binding site is reduced in DDX21SAT. nts, nucleotides. g, qRT–PCR analysis assessing the expression levels of several snoRNAs 24 h after expression of either DDX21WT or DDX21SAT. This experiment was performed in biological duplicates. Data are mean and s.d. of technical triplicates.

Extended Data Figure 7 DDX21 functionally interacts with snoRNAs and the snoRNP.

a, UCSC genome browser view of DDX21WT iCLIP reads across the snorD66 snoRNA. The C box [C] and D box [D] regions are highlighted in red. b, Same visualization as in a but showing the snorA67 snoRNA with the H box [H] and ACA box [ACA] regions highlighted. c, Immunoprecipitation of NOP58 from HEK293 nuclear extracts confirms DDX21 as a protein member of the snoRNP machinery. As a control for this experiment we performed western blots against FBL, a well-known NOP58-interacting partner and an essential factor of the snoRNP machinery. d, DDX21 interacts with XRN2, a 5′–3′ exoribonuclease required for maturation and processing of snoRNAs. The DDX21–XRN2 interaction appears to be bridged by RNA, as treatment of the nuclear lysates with RNaseA abolishes the interaction. e, Schematic of the site-directed RNaseH cleavage of RNA sensitive to 2′-Ome. RNA of interest is hybridized to a 2′-Ome/DNA chimaeric oligonucleotide in which the DNA nucleotides specifically target the ability of RNaseH to interrogate the 2′-Ome status of a single nucleotide. 2′-Ome will inhibit RNaseH and leave intact RNA, while unmethylated RNA will be cleaved. f, UCSC genome browser view of DDX21WT and DDX21SAT iCLIP reads across the snoRNAs responsible for guiding the modifications tested in Fig. 3f.

Extended Data Figure 8 Association of DDX21 with the RNA and protein components of the 7SK snRNP.

a, Comparison of DDX21WT ChIP-seq targets to DDX21WT iCLIP-bound mRNAs. The numbers of unique and common genes are represented, revealing that most ChIP-seq-bound genes are not immunoprecipitated by iCLIP but that some are recovered in both assays. b, iCLIP read distribution of DDX21WT-target mRNAs categorized by the regions within mRNAs that were bound. Most iCLIP reads fell outside the 5′ UTR. c, DDX21WT iCLIP reads mapping to short repetitive RNAs of the human genome. Percentages of the top four short repetitive RNAs are shown. d, Secondary structure model of the 7SK snRNA annotated with iCLIP reverse transcription stops identified from the DDX21WT (blue) and DDX21SAT (orange) experiments. Nucleotides commonly crosslinked are labelled in green. Known RNA binding protein sites: HEXIM1/2 is highlighted in purple; P-TEFb is highlighted in red; and other P-TEFb ‘release’ factors in the centre are highlighted in green. e, Co-immunoprecipitation analysis of HEXIM1 as assayed by western blotting for DDX21WT and 7SK snRNP components (CDK9 and LARP7). f, Immunoprecipitation of Flag–HA–DDX21WT from HEK293 nuclear extracts confirms DDX21 as a protein component of the 7SK snRNP through co-recovery of LARP7. The abundant protein actin, which is not part of the 7SK snRNP, was not recovered.

Extended Data Figure 9 Binding of the DDX21–7SK snRNP at ribosomal gene promoters.

a, b, ChIP-qPCR of CDK9 (a) and HEXIM1 (b) in HEK293 cells at representative Pol II-regulated, DDX21-target and -non-target gene promoters, negative control regions, and the rDNA locus. c, ChIP-qPCR of DDX21 in control or 3′-7SK-ASO-treated HEK293 cells at representative Pol II-regulated, DDX21-target gene promoters, negative control regions, and the rDNA locus. For the promoter-associated genes, P ≤ 0.05 (Student’s t-test) when compared to control ASO. d, Gene Ontology molecular function and cellular component analysis of publically available CDK9 ChIP-seq data30. e, ChIP-qPCR of total Pol II in control or DDX21-targeting siRNA-treated HEK293 cells at representative TSSs of Pol II-regulated, DDX21-target gene promoters and negative control regions. Data are mean and s.d. of three independent experiments.

Extended Data Figure 10 Catalytically inactive DDX21 is still incorporated into the 7SK snRNP.

a, ChIP-qPCR of DDX21WT (black) and DDX21SAT (green) in HEK293 cells at representative TSSs of Pol II-regulated, DDX21-target gene promoters and negative control regions. Data are mean and s.d. of three independent experiments. b, Immunoprecipitation of DDX21WT, DDX21DEV or DDX21SAT from HEK293 nuclear extracts confirms DDX21 interacts with CDK9 (P-TEFb) regardless of its catalytic activity. c, DDX21WT (blue) and DDX21SAT (orange) annotated iCLIP reads mapped across the 7SK snRNA. The four annotated stem–loops are marked below the graph. d, Model of multi-level control of ribosomal pathway by DDX21. In the nucleolus, DDX21 associates with the chromatin across the transcribed region of the rDNA and is a component of the snoRNP. Furthermore, DDX21 functionally interacts with the rRNA, snoRNAs and snoRNP to control 2′-Ome deposition on the rRNA in a helicase activity-dependent manner. In the nucleoplasm, DDX21 is bound to the promoter regions of ribosomal Pol II-transcribed genes, many of which contain precursor snoRNA transcripts. Mechanistically, DDX21 activates transcription of its target genes through the 7SK–P-TEFb axis. As part of the 7SK snRNP, DDX21 can facilitate the release of P-TEFb from the inhibitory complex in a manner dependent on ATP hydrolysis, leading to productive Pol II elongation and increased phosphorylation of Ser 2. Efficient transcription of its target genes enforces high expression of both snoRNAs and other ribosomal proteins critical for the rRNA maturation process, placing DDX21 as a central operator of the ribosomal pathway.

Supplementary information

Supplementary Table I

This fie contains oligonucleotides utilized in this study. (XLSX 51 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Calo, E., Flynn, R., Martin, L. et al. RNA helicase DDX21 coordinates transcription and ribosomal RNA processing. Nature 518, 249–253 (2015). https://doi.org/10.1038/nature13923

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13923

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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