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:

Mapping the genomic landscape of CRISPR–Cas9 cleavage

An Author Correction to this article was published on 09 November 2023

This article has been updated

Abstract

RNA-guided CRISPR–Cas9 endonucleases are widely used for genome engineering, but our understanding of Cas9 specificity remains incomplete. Here, we developed a biochemical method (SITE-Seq), using Cas9 programmed with single-guide RNAs (sgRNAs), to identify the sequence of cut sites within genomic DNA. Cells edited with the same Cas9–sgRNA complexes are then assayed for mutations at each cut site using amplicon sequencing. We used SITE-Seq to examine Cas9 specificity with sgRNAs targeting the human genome. The number of sites identified depended on sgRNA sequence and nuclease concentration. Sites identified at lower concentrations showed a higher propensity for off-target mutations in cells. The list of off-target sites showing activity in cells was influenced by sgRNP delivery, cell type and duration of exposure to the nuclease. Collectively, our results underscore the utility of combining comprehensive biochemical identification of off-target sites with independent cell-based measurements of activity at those sites when assessing nuclease activity and specificity.

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: SITE-Seq workflow.
Figure 2: SITE-Seq target sites recovered with 0.25–1,024 nM Cas9.
Figure 3: SITE-Seq predicts cell-based activity.
Figure 4: Off-target editing varies with sgRNP delivery method, duration of treatment and cell type.
Figure 5: Comparison of SITE-Seq with other off-target analysis methods.

Similar content being viewed by others

Accession codes

Primary accessions

BioProject

Change history

References

  1. Hsu, P.D., Lander, E.S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  PubMed  Google Scholar 

  3. Sternberg, S.H. & Doudna, J.A. Expanding the biologist's toolkit with CRISPR-Cas9. Mol. Cell 58, 568–574 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gasiunas, G., Barrangou, R., Horvath, P. & Siksnys, V. Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl. Acad. Sci. USA 109, E2579–E2586 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Anders, C., Niewoehner, O., Duerst, A. & Jinek, M. Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature 513, 569–573 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Nishimasu, H. et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell 156, 935–949 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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  PubMed  PubMed Central  Google Scholar 

  10. 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  PubMed  PubMed Central  Google Scholar 

  11. Tsai, S.Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Frock, R.L. et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat. Biotechnol. 33, 179–186 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Kim, D. et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells. Nat. Methods 12, 237–243, 1 p following 243 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Kim, D., Kim, S., Kim, S., Park, J. & Kim, J.-S. Genome-wide target specificities of CRISPR-Cas9 nucleases revealed by multiplex Digenome-seq. Genome Res. 26, 406–415 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fu, B.X.H., St Onge, R.P., Fire, A.Z. & Smith, J.D. Distinct patterns of Cas9 mismatch tolerance in vitro and in vivo. Nucleic Acids Res. 44, 5365–5377 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Fu, B.X.H., Hansen, L.L., Artiles, K.L., Nonet, M.L. & Fire, A.Z. Landscape of target:guide homology effects on Cas9-mediated cleavage. Nucleic Acids Res. 42, 13778–13787 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Wang, X. et al. Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors. Nat. Biotechnol. 33, 175–178 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Crosetto, N. et al. Nucleotide-resolution DNA double-strand break mapping by next-generation sequencing. Nat. Methods 10, 361–365 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kim, S., Kim, D., Cho, S.W., Kim, J. & Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012–1019 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Liang, X. et al. Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J. Biotechnol. 208, 44–53 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Richardson, C.D., Ray, G.J., Bray, N.L. & Corn, J.E. Non-homologous DNA increases gene disruption efficiency by altering DNA repair outcomes. Nat. Commun. 7, 12463 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Horlbeck, M.A. et al. Nucleosomes impede Cas9 access to DNA in vivo and in vitro. eLife 5, e12677 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Boyle, A.P., Guinney, J., Crawford, G.E. & Furey, T.S. F-Seq: a feature density estimator for high-throughput sequence tags. Bioinformatics 24, 2537–2538 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Stemmer, M., Thumberger, T., Del Sol Keyer, M., Wittbrodt, J. & Mateo, J.L. CCTop: an intuitive, flexible and reliable CRISPR/Cas9 target prediction tool. PLoS One 10, e0124633 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Bae, S., Park, J. & Kim, J.-S. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 30, 1473–1475 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Cameron, P. et al. SITE-Seq: a genome-wide method to measure Cas9 cleavage. Protocol Exchange http://dx.doi.org/10.1038/protex.2017.043 (2017).

  28. Briner, A.E. et al. Guide RNA functional modules direct Cas9 activity and orthogonality. Mol. Cell 56, 333–339 (2014).

    Article  CAS  PubMed  Google Scholar 

  29. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Bailey, T.L. & Elkan, C. Fitting a mixture model by expectation maximization to discover motifs in biopolymers. In Proc. Int. Conf. Intell. Syst. Mol. Biol. (eds. Altman, R. et al.) 28–36 (AAAI Press, 1994 ).

  33. Blitzblau, H.G., Bell, G.W., Rodriguez, J., Bell, S.P. & Hochwagen, A. Mapping of meiotic single-stranded DNA reveals double-stranded-break hotspots near centromeres and telomeres. Curr. Biol. 17, 2003–2012 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Capes-Davis, A. et al. Check your cultures! A list of cross-contaminated or misidentified cell lines. Int. J. Cancer 127, 1–8 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank R. Haurwitz, S. Sternberg, B. McClung, and the members of Caribou Biosciences for providing helpful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

J.K.Y., V.L., S.D., M.C., A.P.M. and P.D.D. designed critical proof-of-concept experiments to initially develop the SITE-Seq protocol. P.C. performed all SITE-Seq experiments in this study; B.N.J. performed amplicon digestion experiments; M.S.T. and L.M.B. performed sequencing for cell-based validation experiments. P.C., E.M.S., B.V., E.G. and E.L. performed cell-based validation experiments. S.G., A.M.L. and L.S.E. generated sgRNA and other reagents. P.C., C.K.F., P.D.D., M.M.C., D.C., M.C., B.N.J., A.H.S., J.K.Y. and A.P.M. analyzed the data. P.C., C.K.F., P.D.D., M.C., J.K.Y. and A.P.M. wrote the manuscript.

Corresponding authors

Correspondence to Joshua K Young or Andrew P May.

Ethics declarations

Competing interests

A.P.M., P.D.D., P.C., C.K.F., M.S.T., M.M.C., S.G., B.N.J., E.L., E.M.S., B.V., E.L., L.M.B., A.M.L., L.S.E., A.H.S., and D.C. are current or former employees of Caribou Biosciences, Inc., a company that develops and commercializes genome engineering technologies; and such individuals may own shares or stock options in Caribou Biosciences, Inc. J.K.Y., V.L., and S.D. are employees of DuPont Pioneer. Patent applications have been filed describing this methodology; see, for example, PCT publication no. WO2014/164466, published October 9, 2014.

Integrated supplementary information

Supplementary Figure 1 Sequence logos resulting from SITE-Seq target sites recovered with 0.25 nM – 1,024 nM sgRNP.

The total number of SITE-Seq target sites recovered at each sgRNP concentration is displayed (upper left corner) with each logo. On-target sequences are directly below the gene name. The concentration of sgRNP used in SITE-Seq is shown to the right.

Supplementary Figure 2 SITE-Seq target sites with higher biochemical cleavage sensitivities are more likely to show cell-based editing.

(a) For each sgRNA, the fraction of SITE-Seq target sites showing cell-based editing as a function of SITE-Seq digestion conditions. (b) Across all sites examined in cells, the fraction of SITE-Seq target sites showing cell-based editing as a function of SITE-Seq digestion conditions.

Supplementary Figure 3 DNase-I hypersensitivity does not correlate with on- or off-target activity.

DNase-I hypersensitivity does not correlate with on- or off-target activity. (a) Scatterplots of indel frequencies at on- and -off-target sites as a function of enrichment scores for DNase-I hypersensitivity density (generated by F-Seq). sgRNAs targeting AAVS1, VEGFA, or FANCF were transfected into HEK293 cells stably expressing Cas9-GFP and on- and off-target editing was measured three days later. (b) Scatterplots of indel frequencies at SITE-Seq target sites directly targeted with sgRNAs as a function of enrichment scores for DNase-I hypersensitivity density (generated by F-Seq). 387 sgRNAs were transfected into HEK293 cells stably expressing Cas9-GFP and on-target editing at each SITE-Seq target site was measured two days later.

Supplementary Figure 4 SITE-Seq mediated sgRNA selection.

(a) The EGFR gene was targeted with 83 sgRNAs tiling across six exons. Shown is the full EGFR locus as well as the first six exons and their corresponding guides (grey guides target sense strand and red guides target anti-sense strand). (b) Overlap between SITE-Seq target sites recovered with 4 nM positive control sgRNPs and cellular off-targets observed after transient transfection of pre-assembled sgRNP. (c) Dot plots show coupling between EGFR protein knockdown and recovery of SITE-Seq target sites. sgRNAs with high on-target activity (>70% EGFR knockdown) and less than 30 SITE-Seq target sites are shown as either orange (if at least one SITE-Seq target site is found in an exon) or green (if no SITE-Seq target site is found in an exon).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 (PDF 792 kb)

Supplementary Table 1–9

SITE-Seq target sites recovered (XLSX 1207 kb)

Supplementary Table 10

SITE-Seq target sites recovered with VEGFA sgRNP that were segregated by MEME into a second motif. (XLSX 114 kb)

Supplementary Table 11

Biochemical cleavage of amplicons containing SITE-Seq target sites, as a function of sgRNP concentration. (XLSX 14 kb)

Supplementary Table 12–19

SITE-Seq target sites examined in cell-based validation. (XLSX 208 kb)

Supplementary Table 20–22

SITE-Seq target sites examined in cell-based validation with a panel of delivery methods. (XLSX 71 kb)

Supplementary Table 23

SITE-Seq with high sequencing coverage. (XLSX 62 kb)

Supplementary Table 24

Comparing SITE-Seq data with in silico approaches (XLSX 9 kb)

Supplementary Table 25-26

Oligonucleotides used in SITE-Seq. (XLSX 11 kb)

Supplementary Protocol

SITE-Seq Supplementary Protocol (PDF 1253 kb)

Supplementary Software

SITE-Seq feature calling function (TXT 8 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cameron, P., Fuller, C., Donohoue, P. et al. Mapping the genomic landscape of CRISPR–Cas9 cleavage. Nat Methods 14, 600–606 (2017). https://doi.org/10.1038/nmeth.4284

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.4284

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