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.

  • Technical Report
  • Published:

Recombinant Dicer efficiently converts large dsRNAs into siRNAs suitable for gene silencing

Abstract

RNA interference (RNAi) is a powerful method for specifically silencing gene expression in diverse cell types1,2,3. RNAi is mediated by 21-nucleotide small interfering RNAs (siRNAs)4,5,6,7,8, which are produced from larger double-stranded RNAs (dsRNAs) in vivo through the action of Dicer, an RNase III–family enzyme9,10,11. Transfecting cells with siRNAs rather than larger dsRNAs avoids the nonspecific gene silencing of the interferon response12, underscoring the importance of developing efficient methods for producing reliable siRNAs. Here we show that pools of 20- to 21-base pair (bp) siRNAs can be produced enzymatically in vitro using active recombinant Dicer. Yields of ≤ 70% are obtained, and the siRNAs can be easily separated from any residual large dsRNA by a series of spin columns or gel purification. Dicer-generated siRNAs (d-siRNAs) are effective in silencing transiently transfected reporter genes and endogenous genes, making in vitro dicing a useful, practical alternative for the production of siRNAs.

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: Production of pools of siRNAs by purified recombinant Dicer.
Figure 2: d-siRNAs specifically silence luciferase expression in HEK 293 cells.
Figure 3: d-siRNAs can silence endogenous genes.

Similar content being viewed by others

References

  1. Fire, A. RNA-triggered gene silencing. Trends Genet. 15, 358–363 (1999).

    Article  CAS  Google Scholar 

  2. Sharp, P.A. RNA interference—2001. Genes Dev. 15, 485–490 (2001).

    Article  CAS  Google Scholar 

  3. Paddison, P.J. & Hannon, G.J. RNA interference: the new somatic cell genetics? Cancer Cell 2, 17–23 (2002).

    Article  CAS  Google Scholar 

  4. Zamore, P.D., Tuschl, T., Sharp, P.A. & Bartel, D.P. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21- to 23-nucleotide intervals. Cell 101, 25–33 (2000).

    Article  CAS  Google Scholar 

  5. Caplen, N.J., Parrish, S., Imani, F., Fire, A. & Morgan, R.A. Specific inhibition of gene expression by small double-stranded RNAs in invertebrates and vertebrate systems. Proc. Natl. Acad. Sci. USA 98, 9742–9747 (2001).

    Article  CAS  Google Scholar 

  6. Elbashir, S.M. et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411, 494–498 (2001).

    Article  CAS  Google Scholar 

  7. Elbashir, S.M., Lendeckel, W. & Tuschl, T. RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes Dev. 15, 188–200 (2001).

    Article  CAS  Google Scholar 

  8. Elbashir, S.M., Martinez, J., Patkaniowska, A., Lendeckel, W. & Tuschl, T. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J. 20, 6877–6888 (2001).

    Article  CAS  Google Scholar 

  9. Bernstein, E., Caudy, A.A., Hammond, S.M. & Hannon, G.J. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 409, 363–366 (2001).

    Article  CAS  Google Scholar 

  10. Ketting, R.F. et al. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev. 15, 2654–2659 (2001).

    Article  CAS  Google Scholar 

  11. Knight, S.W. & Bass, B.L. A role for the RNase III enzyme DCR-1 in RNA interference and germ line development in Caenorhabditis elegans. Science 293, 2269–2271 (2001).

    Article  CAS  Google Scholar 

  12. Stark, G.R., Kerr, I.M., Williams, B.R., Silverman, R.H. & Schreiber, R.D. How cells respond to interferons. Annu. Rev. Biochem. 67, 227–264 (1998).

    Article  CAS  Google Scholar 

  13. Yu, J.Y., DeRuiter, S.L. & Turner, D.L. RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells. Proc. Natl. Acad. Sci. USA 99, 6047–6052 (2002).

    Article  CAS  Google Scholar 

  14. Miyagishi, M. & Taira, K. U6 promoter-driven siRNAs with four uridine 3′ overhangs efficiently suppress targeted gene expression in mammalian cells. Nat. Biotechnol. 20, 497–500 (2002).

    Article  CAS  Google Scholar 

  15. Paul, C.P., Good, P.D., Winder, I. & Engelke, D.R. Effective expression of small interfering RNA in human cells. Nat. Biotechnol. 20, 505–508 (2002).

    Article  CAS  Google Scholar 

  16. Lee, N.S. et al. Expression of small interfering RNAs targeted against HIV-1 rev transcripts in human cells. Nat. Biotechnol. 20, 500–505 (2002).

    Article  CAS  Google Scholar 

  17. Brummelkamp, T.R., Bernards, R. & Agami, R. A system for stable expression of short interfering RNAs in mammalian cells. Science 296, 550–553 (2002).

    Article  CAS  Google Scholar 

  18. Yang, D. et al. Short RNA duplexes produced by hydrolysis with Escherichia coli RNase III mediate effective RNA interference in mammalian cells. Proc. Natl. Acad. Sci. USA 99, 9942–9947 (2002).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. Zhang, H., Kolb, F.A., Brondani, V., Billy, E. & Filipowicz, W. Human Dicer preferentially cleaves dsRNAs at their termini without a requirement for ATP. EMBO J. 21, 5875–5885 (2002).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Greg Hannon for providing a CMV-Dicer clone and synthetic siRNAs and for helpful discussions, Thomas Wehrman for help with luciferase assays, M.-C. Yee for help with Hi5 and Sf9 cultures, Jianbo Yue for help with chromatography, and members of the Ferrell lab for advice and critical reading of the manuscript. This work was supported by grants from the National Institutes of Health to J.E.F. (GM46383) and T.M. (CA83229).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jason W. Myers.

Ethics declarations

Competing interests

J.W.M. and J.E.F. Jr. have filed a patent application related to using recombinant Dicer to generate siRNAs as described in this article.

Supplementary information

Supplementary Figure 1.

We expressed full-length, His6- and T7-tagged-Dicer in Hi5 insect cells by baculovirus infection and purified it on a cobalt Sepharose column. Fractions were collected and elecrophoresed in a 6% (wt/vol) SDS-polyacrylamide gel and proteins were detected by either coomassie-staining or immunoblotting. The main protein eluted was a 225 kDa protein, which corresponds to the predicted molecular mass for Dicer (Supplementary Fig. 1A, top left panel). The main immunoreactive band on an anti-tag immunoblot also migrates at the predicted molecular mass for Dicer (Supplementary Fig. 1A, left middle panel), however the 225 kDa band is not present in the elution fractions collected from a mock purification of uninfected cellular lysates in either the coomassie stained gel (Supplementary Fig. 1A, top right panel) or on the immunoblot (Supplementary Fig. 1A, right middle panel). Taken together these results confirm the identity of the 225 kDa band as recombinant Dicer. We then determined whether the purified recombinant Dicer was able to process dsRNA into siRNAs. We incubated column fractions with a 500 bp dsRNA and analyzed the reaction products by native polyacrylamide gel electrophoresis. The purified Dicer efficiently converted the dsRNA to a form (Supplementary Fig. 1A, bottom left panel) that co-migrated with a synthetic 21 bp siRNA (data not shown). Because Drosophila extracts are capable of generating siRNAs4, it is likely that the Hi5 insect cells used to produce r-Dicer contained some endogenous dicing activity. Therefore, we addressed the question of whether the Dicer activity in our purified fractions was due to the r-Dicer or to some adventitiously co-purifying insect protein. We subjected lysates from uninfected Hi5 cells to the same purification procedure and assessed fractions for Dicer activity. A small amount of RNA processing activity could be detected in the total soluble Hi5 cell lysate and in the flow through from the cobalt affinity column (Supplementary Fig. 1A, bottom right panel). However this activity was not detected in the column eluates, indicating that it could not account for the high levels of dicing activity seen in the purified r-Dicer preparation. In addition, Dicer protein and activities co-chromatographed on Q-Sepharose (Supplementary Fig. 1B), again supporting the hypothesis that the r-Dicer is required for the dicing activity seen. r-Dicer was subjected to various reaction conditions to determine cofactor and substrate specificity as well as to ensure that the 21-mer was generated by an RNase IIIfamily enzyme (like r-Dicer) and not a nonspecific RNase or nuclease. dsRNA was efficiently processed by r-Dicer (Supplementary Fig. 1C, lane 1) but dsDNA was not (Supplementary Fig. 1C, lane 2), establishing that r-Dicer is RNA-specific. r-Dicer generated small amounts of 21 bp product from ssRNA, which may be due to cleavage of hairpins or contaminating dsRNA (Supplementary Fig. 1C, lane 3, and data not shown). r-Dicer was not inhibited by RNasin (Supplementary Fig. 1C, lane 4), distinguishing it from most ribonucleases. Mg2+ was required for r-Dicer activity (Supplementary Fig. 1C, lanes 6 and 7), but ATP was not (Supplementary Fig. 1C, lane 5). In contrast, other workers have found that ATP is important for RNAi and dicing in drosophila extracts4 and in Dicer immunoprecipitates9. However, since submission of this manuscript others have shown that in vitro dicing, at lest with recombinant human Dicer, does not require ATP19,21. High concentrations of manganese (2.5-5 mM) and calcium (10 mM) inhibited r-Dicer activity (data not shown). (PDF 118 kb)

Supplementary Figure 2.

Various 500-2300 bp dsRNAs were cleaved by r-Dicer to generate d-siRNAs. Large dsRNAs shown in Supplementary Figure 2A were 500-600 bps and those shown in Supplementary Figure 2B were 1400-2300 bps (see Supplementary Table 1 online for exact sizes). The contaminating reaction components and larger dsRNAs were separated from the d-siRNAs by a series of spin columns or by gel purification as inidicated. Shown are 15% native polyacrylamide gels after ethidium staining illustrating the quantity, quality and purity of siRNAs and d-siRNAs after purification as well as the constituents of the unpurified reaction. Electrophoresis verified that the d-siRNAs were not contaminated with large dsRNAs, the d-siRNAs remained double stranded, and the concentration estimates (estimated by A260) were accurate. The siRNAs/d-siRNAs shown in Supplementary Figure 2A were used in the experiments shown in Figure 2E-H and those shown in Supplementary Figure 2B were used in the experiments shown in Figure 3B. (PDF 406 kb)

Supplementary Table 1 (PDF 13 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Myers, J., Jones, J., Meyer, T. et al. Recombinant Dicer efficiently converts large dsRNAs into siRNAs suitable for gene silencing. Nat Biotechnol 21, 324–328 (2003). https://doi.org/10.1038/nbt792

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt792

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