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:

Improving CRISPR-Cas nuclease specificity using truncated guide RNAs

This article has been updated

Abstract

Clustered, regularly interspaced, short palindromic repeat (CRISPR) RNA-guided nucleases (RGNs) are highly efficient genome editing tools1,2,3. CRISPR-associated 9 (Cas9) RGNs are directed to genomic loci by guide RNAs (gRNAs) containing 20 nucleotides that are complementary to a target DNA sequence. However, RGNs can induce mutations at sites that differ by as many as five nucleotides from the intended target4,5,6. Here we report that truncated gRNAs, with shorter regions of target complementarity <20 nucleotides in length, can decrease undesired mutagenesis at some off-target sites by 5,000-fold or more without sacrificing on-target genome editing efficiencies. In addition, use of truncated gRNAs can further reduce off-target effects induced by pairs of Cas9 variants that nick DNA (paired nickases). Our results delineate a simple, effective strategy to improve the specificities of Cas9 nucleases or paired nickases.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: On-target genome editing activities of truncated gRNAs and Cas9 nuclease in human cells.
Figure 2: tru-gRNAs exhibit enhanced specificities and function efficiently with Cas9 nuclease and paired Cas9 nickases in human cells.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

Change history

  • 29 January 2014

    In the version of this article initially published online, two nearly identical versions of a sentence were included in the abstract (“In addition, use of truncated gRNAs can further reduce off-target effects induced by pairs of Cas9 variants that nick DNA (paired nickases). Also, use of truncated gRNAs can lead to further reductions in off-target mutations induced by pairs of Cas9 variants that nick DNA (paired nickases).”) and the name and reference citation for a plasmid in the Online Methods was incorrect. The correct sentence is “In addition, use of truncated gRNAs can further reduce off-target effects induced by pairs of Cas9 variants that nick DNA (paired nickases)” and the plasmid name and reference are “pJDS246 (ref. 4),” not “pJDS2464.” These errors have been corrected for the print, PDF and HTML versions of this article.

References

  1. Wiedenheft, B., Sternberg, S.H. & Doudna, J.A. RNA-guided genetic silencing systems in bacteria and archaea. Nature 482, 331–338 (2012).

    Article  CAS  Google Scholar 

  2. Mali, P., Esvelt, K.M. & Church, G.M. Cas9 as a versatile tool for engineering biology. Nat. Methods 10, 957–963 (2013).

    Article  CAS  Google Scholar 

  3. Charpentier, E. & Doudna, J.A. Biotechnology: rewriting a genome. Nature 495, 50–51 (2013).

    Article  CAS  Google Scholar 

  4. Fu, Y. et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 31, 822–826 (2013).

    Article  CAS  Google Scholar 

  5. Hsu, P.D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  Google Scholar 

  6. Pattanayak, V. et al. High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat. Biotechnol. 31, 839–843 (2013).

    Article  CAS  Google Scholar 

  7. Cho, S.W. et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. 24, 132–141 (2014).

    Article  CAS  Google Scholar 

  8. Kim, E. et al. Precision genome engineering with programmable DNA-nicking enzymes. Genome Res. 22, 1327–1333 (2012).

    Article  CAS  Google Scholar 

  9. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838 (2013).

    Article  CAS  Google Scholar 

  10. Ran, F.A. et al. Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 154, 1380–1389 (2013).

    Article  CAS  Google Scholar 

  11. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  Google Scholar 

  12. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  CAS  Google Scholar 

  13. Jao, L.E., Wente, S.R. & Chen, W. Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proc. Natl. Acad. Sci. USA 110, 13904–13909 (2013).

    Article  CAS  Google Scholar 

  14. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  Google Scholar 

  15. Wang, T., Wei, J.J., Sabatini, D.M. & Lander, E.S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  Google Scholar 

  16. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  Google Scholar 

  17. Gilbert, L.A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    Article  CAS  Google Scholar 

  18. Maeder, M.L. et al. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10, 977–979 (2013).

    Article  CAS  Google Scholar 

  19. Perez-Pinera, P. et al. RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat. Methods 10, 973–976 (2013).

    Article  CAS  Google Scholar 

  20. Hwang, W.Y. et al. Heritable and precise zebrafish genome editing using a CRISPR-Cas System. PLoS ONE 8, e68708 (2013).

    Article  CAS  Google Scholar 

  21. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  Google Scholar 

  22. Reyon, D. et al. FLASH assembly of TALENs for high-throughput genome editing. Nat. Biotechnol. 30, 460–465 (2012).

    Article  CAS  Google Scholar 

  23. Beerli, R.R., Dreier, B. & Barbas, C.F. III. Positive and negative regulation of endogenous genes by designed transcription factors. Proc. Natl. Acad. Sci. USA 97, 1495–1500 (2000).

    Article  CAS  Google Scholar 

  24. Pattanayak, V., Ramirez, C.L., Joung, J.K. & Liu, D.R. Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat. Methods 8, 765–770 (2011).

    Article  CAS  Google Scholar 

  25. Qi, L.S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  Google Scholar 

  26. Sander, J.D. et al. ZiFiT (Zinc Finger Targeter): an updated zinc finger engineering tool. Nucleic Acids Res. 38, W462–W468 (2010).

    Article  CAS  Google Scholar 

  27. Sander, J.D., Zaback, P., Joung, J.K., Voytas, D.F. & Dobbs, D. Zinc Finger Targeter (ZiFiT): an engineered zinc finger/target site design tool. Nucleic Acids Res. 35, W599–W605 (2007).

    Article  Google Scholar 

  28. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  Google Scholar 

  29. Fisher, S. et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 12, R1 (2011).

    Article  Google Scholar 

  30. Sander, J.D. et al. In silico abstraction of zinc finger nuclease cleavage profiles reveals an expanded landscape of off-target sites. Nucleic Acids Res. 41, e181 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Maeder, S. Tsai and J. Angstman for helpful discussions and comments on the manuscript and J. Foden for technical assistance. This work was funded by a National Institutes of Health (NIH) Director's Pioneer Award (DP1 GM105378), NIH R01 GM088040, NIH P50 HG005550, and the Jim and Ann Orr Massachusetts General Hospital (MGH) Research Scholar Award. This material is based upon work supported by, or in part by, the US Army Research Laboratory and the US Army Research Office under grant number W911NF-11-2-0056.

Author information

Authors and Affiliations

Authors

Contributions

Y.F., J.D.S., D.R. and J.K.J. conceived of and designed experiments. Y.F., J.D.S. and V.M.C. performed experiments. D.R. developed the updated version of ZiFiT software. Y.F., J.D.S. and J.K.J. wrote the paper.

Corresponding authors

Correspondence to Jeffry D Sander or J Keith Joung.

Ethics declarations

Competing interests

J.K.J. has financial interests in Editas Medicine and Transposagen Biopharmaceuticals. J.K.J.'s interests were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. J.K.J. and J.D.S. have filed a patent application on the tru-gRNA/tru-RGN technology. J.K.J. and J.D.S. are consultants for Editas Medicine, a company focused on developing genome-editing therapeutics. Following the completion of this work, D.R. has become an employee of Biogen Idec.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Figures 1–6 and Supplementary Tables 1–6 (PDF 1458 kb)

Supplementary Table 7

Primers used to construct gRNA expression vectors (XLSX 26 kb)

Supplementy Table 8

Primers used to amplify genomic loci for T7EI and deep sequencing assays (XLSX 62 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fu, Y., Sander, J., Reyon, D. et al. Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol 32, 279–284 (2014). https://doi.org/10.1038/nbt.2808

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.2808

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research