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:

A biologically stable DNAzyme that efficiently silences gene expression in cells

Matters Arising to this article was published on 18 July 2022

Abstract

Efforts to use RNA-cleaving DNA enzymes (DNAzymes) as gene-silencing agents in therapeutic applications have stalled due to their low efficacy in clinical trials. Here we report a xeno-nucleic-acid-modified version of the classic DNAzyme 10–23 that achieves multiple-turnover activity under cellular conditions and resists nuclease digestion. The new reagent, X10–23, overcomes the problem of product inhibition, which limited previous 10–23 designs, using molecular chemotypes with DNA, 2′-fluoroarabino nucleic acid and α-l-threofuranosyl nucleic acid backbone architectures that balance the effects of enhanced biological stability with RNA hybridization and divalent metal ion coordination. In cultured mammalian cells, X10–23 facilitates persistent gene silencing by efficiently degrading exogenous and endogenous messenger RNA transcripts. Together, these results demonstrate that new molecular chemotypes can improve the activity and stability of DNAzymes, and may provide a new route for nucleic acid enzymes to reach the clinic.

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: Kinetic analysis of F10–23.
Fig. 2: Engineering of the X10–23 nucleic acid enzyme.
Fig. 3: Functional activity and biostability of X10–23.
Fig. 4: Alternative 10–23 designs.
Fig. 5: GFP inhibition activity of X10–23 in HEK293 cells.
Fig. 6: Targeting endogenous oncogene KRAS by X10–23 in cancer cells.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available within the article and its Supplementary Information files. Source data are provided with this paper.

References

  1. Santoro, S. W. & Joyce, G. F. A general purpose RNA-cleaving DNA enzyme. Proc. Natl Acad. Sci. USA 94, 4262–4266 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Joyce, G. F. RNA cleavage by the 10–23 DNA enzyme. Methods Enzymol. 341, 503–517 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Khvorova, A. & Watts, J. K. The chemical evolution of oligonucleotide therapies of clinical utility. Nat. Biotechnol. 35, 238–248 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Santoro, S. W. & Joyce, G. F. Mechanism and utility of an RNA-cleaving DNA enzyme. Biochemistry 37, 13330–13342 (1998).

    Article  CAS  PubMed  Google Scholar 

  5. Appaiahgari, M. B. & Vrati, S. DNAzyme-mediated inhibition of Japanese encephalitis virus replication in mouse brain. Mol. Ther. 15, 1593–1599 (2007).

    Article  CAS  PubMed  Google Scholar 

  6. Xie, Y. Y. et al. Inhibition of respiratory syncytial virus in cultured cells by nucleocapsid gene targeted deoxyribozyme (DNAzyme). Antivir. Res. 71, 31–41 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Schubert, S. et al. RNA cleaving ‘10–23′ DNAzymes with enhanced stability and activity. Nucleic Acids Res. 31, 5982–5992 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fokina, A. A., Meschaninova, M. I., Durfort, T., Venyaminova, A. G. & Francois, J. C. Targeting insulin-like growth factor I with 10–23 DNAzymes: 2′-O-methyl modifications in the catalytic core enhance mRNA cleavage. Biochemistry 51, 2181–2191 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Takahashi, H. et al. A new modified DNA enzyme that targets influenza virus A mRNA inhibits viral infection in cultured cells. FEBS Lett. 560, 69–74 (2004).

    Article  CAS  PubMed  Google Scholar 

  10. Vester, B. et al. LNAzymes: incorporation of LNA-type monomers into DNAzymes markedly increases RNA cleavage. J. Am. Chem. Soc. 124, 13682–13683 (2002).

    Article  CAS  PubMed  Google Scholar 

  11. Fokina, A. A., Chelobanov, B. P., Fujii, M. & Stetsenko, D. A. Delivery of therapeutic RNA-cleaving oligodeoxyribonucleotides (deoxyribozymes): from cell culture studies to clinical trials. Expert. Opin. Drug Deliv. 14, 1077–1089 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Cho, E. A. et al. Safety and tolerability of an intratumorally injected DNAzyme, Dz13, in patients with nodular basal-cell carcinoma: a phase 1 first-in-human trial (DISCOVER). Lancet 381, 1835–1843 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cao, Y. et al. Therapeutic evaluation of Epstein–Barr virus-encoded latent membrane protein-1 targeted DNAzyme for treating of nasopharyngeal carcinomas. Mol. Ther. 22, 371–377 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Hohlfeld, J. et al. Safety profile and pharmacokinetics of an inhaled GATA-3-specific DNAzyme in a phase Ib study in patients with stable allergic asthma. Eur. Respir. J. 42, 4856 (2013).

    Google Scholar 

  15. Cai, H. et al. DNAzyme targeting c-jun suppresses skin cancer growth. Sci. Transl. Med. 4, 139ra182 (2012).

    Article  Google Scholar 

  16. Dicke, T. et al. Absence of unspecific innate immune cell activation by GATA-3-specific DNAzymes. Nucleic Acid Ther. 22, 117–126 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Chaput, J. C., Herdewijn, P. & Hollenstein, M. Orthogonal genetic systems. ChemBioChem 21, 1408–1411 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Damha, M. J. et al. Hybrids of RNA and arabinonucleic acids (ANA and 2′F-ANA) are substrates for ribonuclease H. J. Am. Chem. Soc. 120, 12976–12977 (1998).

    Article  CAS  Google Scholar 

  19. Wilds, C. J. & Damha, M. J. 2′-Deoxy-2′-fluoro-β-d-arabinonucleosides and oligonucleotides (2′F-ANA): synthesis and physicochemical studies. Nucleic Acids Res. 28, 3625–3635 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jakobsen, M. R., Haasnoot, J., Wengel, J., Berkhout, B. & Kjems, J. Efficient inhibition of HIV-1 expression by LNA modified antisense oligonucleotides and DNAzymes targeted to functionally selected binding sites. Retrovirology 4, 29 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Wolf, F. I., Torsello, A., Fasanella, S. & Cittadini, A. Cell physiology of magnesium. Mol. Aspects Med. 24, 11–26 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Jung, D. W., Apel, L. & Brierley, G. P. Matrix free Mg2+ changes with metabolic state in isolated heart mitochondria. Biochemistry 29, 4121–4128 (1990).

    Article  CAS  PubMed  Google Scholar 

  23. Cozens, C. et al. Enzymatic synthesis of nucleic acids with defined regioisomeric 2′–5′ linkages. Angew. Chem. Int. Ed. 54, 15570–15573 (2015).

    Article  CAS  Google Scholar 

  24. Schöning, K. U. et al. Chemical etiology of nucleic acid structure: the ɑ-threofuranosyl-(3′→2′) oligonucleotide system. Science 290, 1347–1351 (2000).

    Article  PubMed  Google Scholar 

  25. Culbertson, M. C. et al. Evaluating TNA stability under simulated physiological conditions. Bioorg. Med. Chem. Lett. 26, 2418–2421 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Barrett, S. E. et al. An in vivo evaluation of amphiphilic, biodegradable peptide copolymers as siRNA delivery agents. Int. J. Pharm. 466, 58–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Cummins, L. L. et al. Characterization of fully 2′-modified oligoribonucleotide hetero- and homoduplex hybridization and nuclease sensitivity. Nucleic Acids Res. 23, 2019–2024 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Prakash, T. P. An overview of sugar-modified oligonucleotides for antisense therapeutics. Chem. Biodivers. 8, 1616–1641 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Farber, S., D’Angio, G., Evans, A. & Mitus, A. Clinical studies on actinomycin D with special reference to Wilms’ tumor in children. Ann. NY Acad. Sci. 89, 421–425 (1960).

    Article  CAS  PubMed  Google Scholar 

  30. Bensaude, O. Inhibiting eukaryotic transcription: which compound to choose? How to evaluate its activity? Transcription 2, 103–108 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Chiosea, S. I., Sherer, C. K., Jelic, T. & Dacic, S. KRAS mutant allele-specific imbalance in lung adenocarcinoma. Mod. Pathol. 24, 1571–1577 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Krasinskas, A. M., Moser, A. J., Saka, B., Adsay, N. V. & Chiosea, S. I. KRAS mutant allele-specific imbalance is associated with worse prognosis in pancreatic cancer and progression to undifferentiated carcinoma of the pancreas. Mod. Pathol. 26, 1346–1354 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hartman, D. J., Davison, J. M., Foxwell, T. J., Nikiforova, M. N. & Chiosea, S. I. Mutant allele-specific imbalance modulates prognostic impact of KRAS mutations in colorectal adenocarcinoma and is associated with worse overall survival. Int. J. Cancer 131, 1810–1817 (2012).

    Article  CAS  PubMed  Google Scholar 

  34. McCormick, F. KRAS as a therapeutic target. Clin. Cancer Res. 21, 1797–1801 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Young, D. D., Lively, M. O. & Deiters, A. Activation and deactivation of DNAzyme and antisense function with light for the photochemical regulation of gene expression in mammalian cells. J. Am. Chem. Soc. 132, 6183–6193 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fokina, A. A., Stetsenko, D. A. & Francois, J. C. DNA enzymes as potential therapeutics: towards clinical application of 10–23 DNAzymes. Expert Opin. Biol. Ther. 15, 689–711 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Hollenstein, M., Hipolito, C. J., Lam, C. H. & Perrin, D. M. Toward the combinatorial selection of chemically modified DNAzyme RNase A mimics active against all-RNA substrates. ACS Comb. Sci. 15, 174–182 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Wang, Y., Liu, E., Lam, C. H. & Perrin, D. M. A densely modified M(2+)-independent DNAzyme that cleaves RNA efficiently with multiple catalytic turnover. Chem. Sci. 9, 1813–1821 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Silverman, S. K. Catalytic DNA: scope, applications, and biochemistry of deoxyribozymes. Trends Biochem. Sci. 41, 595–609 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Chaput, J. C., Yu, H. & Zhang, S. The emerging world of synthetic genetics. Chem. Biol. 19, 1360–1371 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Pinheiro, V. B., Loakes, D. & Holliger, P. Synthetic polymers and their potential as genetic materials. Bioessays 35, 113–122 (2012).

    Article  PubMed  Google Scholar 

  42. Anosova, I. et al. The structural diversity of artificial genetic polymers. Nucleic Acids Res. 44, 1007–1021 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Kim, J. et al. Patient-customized oligonucleotide therapy for a rare genetic disease. N. Engl. J. Med. 381, 1644–1652 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liao, H. et al. Development of allele-specific therapeutic siRNA in Meesmann epithelial corneal dystrophy. PLoS ONE 6, e28582 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Christie, K. A. et al. Towards personalised allele-specific CRISPR gene editing to treat autosomal dominant disorders. Sci. Rep. 7, 16174 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Sau, S. P., Fahmi, N. E., Liao, J.-Y., Bala, S. & Chaput, J. C. A scalable synthesis of α-l-threose nucleic acid monomers. J. Org. Chem. 81, 2302–2307 (2016).

Download references

Acknowledgements

This work was supported by the W. M. Keck Foundation. Y.W. was supported by a postdoctoral fellowship from the Simons Collaboration on the Origins of Life.

Author information

Authors and Affiliations

Authors

Contributions

J.C.C. and R.C.S. conceived the project and designed the experiments. Y.W. and K.N. performed the experiments. J.C.C. wrote the manuscript with drafts from Y.W. and K.N. All the authors reviewed and commented on the manuscript.

Corresponding author

Correspondence to John C. Chaput.

Ethics declarations

Competing interests

The authors and the University of California-Irvine have filed a patent application on the X10–23 reagent.

Additional information

Peer review information Nature Chemistry thanks Yingfu Li, Chuanzheng Zhou and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Mechanistic analysis of X10-23.

Representative gels showing RNA cleavage activity in the presence and absence of RNase H for an internal segment of GFP (a-c) and the first exon segment of KRAS RNA (d-f). Color code: RNA (red), DNA (black), FANA (orange), and TNA (blue). (a,d) X10-23 with an active catalytic core. (b,e) X10-23 with an inactive catalytic core. (c,f) X10-23 with an active catalytic core that does not hybridize to the RNA target. All assays were performed in buffer containing 0.5 mM MgCl2 and 150 mM NaCl at 37 °C (pH 7.5) with 1 μM substrate and 1 μM enzyme. Nuclease reactions included 0.1 unit/μL of RNase H. S: full-length substrate, P: 5’ cleavage product. Molecular weight markers indicated to the right of the gel.

Source data.

Supplementary information

Supplementary Information

Supplementary Figs. 1–11, Tables 1 and 2, and Source Data.

Reporting Summary

Source data

Source Data Fig. 2

Uncropped gel from Fig. 2.

Source Data Fig. 3

Uncropped gels from Fig. 3a.

Source Data Fig. 4

Uncropped gels from Fig. 4c.

Source Data Extended Data Fig. 1

Uncropped gels from Extended Data Fig. 1.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Nguyen, K., Spitale, R.C. et al. A biologically stable DNAzyme that efficiently silences gene expression in cells. Nat. Chem. 13, 319–326 (2021). https://doi.org/10.1038/s41557-021-00645-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41557-021-00645-x

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