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:

DNA stretching induces Cas9 off-target activity

Abstract

CRISPR/Cas9 is a powerful genome-editing tool, but spurious off-target edits present a barrier to therapeutic applications. To understand how CRISPR/Cas9 discriminates between on-targets and off-targets, we have developed a single-molecule assay combining optical tweezers with fluorescence to monitor binding to λ-DNA. At low forces, the Streptococcus pyogenes Cas9 complex binds and cleaves DNA specifically. At higher forces, numerous off-target binding events appear repeatedly at the same off-target sites in a guide-RNA-sequence-dependent manner, driven by the mechanical distortion of the DNA. Using single-molecule Förster resonance energy transfer (smFRET) and cleavage assays, we show that DNA bubbles induce off-target binding and cleavage at these sites, even with ten mismatches, as well as at previously identified in vivo off-targets. We propose that duplex DNA destabilization during cellular processes (for example, transcription, replication, etc.) can expose these cryptic off-target sites to Cas9 activity, highlighting the need for improved off-target prediction algorithms.

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: Cas9 binds and cleaves DNA tightly and specifically at low force.
Fig. 2: DNA stretching induces Cas9 off-target binding.
Fig. 3: Off-target binding is crRNA sequence specific.
Fig. 4: DNA bubbles induce off-target binding and cleavage.

Similar content being viewed by others

Code availability

All custom code is available on request.

Data availability

All data are available in the main text, the supplementary materials, or upon request. Source data for Figs. 2b, 3c and 4f are available with the paper online.

References

  1. Sander, J. D. & Joung, J. K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 32, 347–355 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Liang, P. et al. CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes. Protein Cell 6, 363–372 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Maeder, M. L. & Gersbach, C. A. Genome-editing technologies for gene and cell therapy. Mol. Ther. 24, 430–446 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Makarova, K. S. et al. Evolution and classification of the CRISPR–Cas systems. Nat. Rev. Microbiol. 9, 467–477 (2011).

    CAS  PubMed  Google Scholar 

  5. Kim, K. et al. Highly efficient RNA-guided base editing in mouse embryos. Nat. Biotechnol. 35, 435–437 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gilbert, L. A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Schwank, G. et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell 13, 653–658 (2013).

    CAS  PubMed  Google Scholar 

  10. Kuscu, C., Arslan, S., Singh, R., Thorpe, J. & Adli, M. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat. Biotechnol. 32, 677–683 (2014).

    CAS  PubMed  Google Scholar 

  11. Cameron, P. et al. Mapping the genomic landscape of CRISPR–Cas9 cleavage. Nat. Methods 14, 600–606 (2017).

    CAS  PubMed  Google Scholar 

  12. Duan, J. et al. Genome-wide identification of CRISPR/Cas9 off-targets in human genome. Cell Res. 24, 1009–1012 (2014).

    CAS  PubMed  PubMed Central  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 (2015).

    CAS  PubMed  Google Scholar 

  14. 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).

    CAS  PubMed  Google Scholar 

  15. Tsai, S. Q. et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR–Cas9 nuclease off-targets. Nat. Methods 14, 607–614 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Wu, X. et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 32, 670–676 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Anderson, K. R. et al. CRISPR off-target analysis in genetically engineered rats and mice. Nat. Methods 15, 512–514 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Akcakaya, P. et al. In vivo CRISPR editing with no detectable genome-wide off-target mutations. Nature 561, 416–419 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62–67 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chen, J. S. et al. Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature 550, 407–410 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Szczelkun, M. D. et al. Direct observation of R-loop formation by single RNA-guided Cas9 and cascade effector complexes. Proc. Natl Acad. Sci. USA 111, 9798–9803 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Singh, D., Sternberg, S. H., Fei, J., Doudna, J. A. & Ha, T. Real-time observation of DNA recognition and rejection by the RNA-guided endonuclease Cas9. Nat. Commun. 7, 12778 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Boyle, E. A. et al. High-throughput biochemical profiling reveals sequence determinants of dCas9 off-target binding and unbinding. Proc. Natl Acad. Sci. USA 114, 5461–5466 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Hirano, S., Nishimasu, H., Ishitani, R. & Nureki, O. Structural basis for the altered PAM specificities of engineered CRISPR-Cas9. Mol. Cell 61, 886–894 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Lim, Y. et al. Structural roles of guide RNAs in the nuclease activity of Cas9 endonuclease. Nat. Commun. 7, 13350 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Knight, S. C. et al. Dynamics of CRISPR-Cas9 genome interrogation in living cells. Science 350, 823–826 (2015).

    CAS  PubMed  Google Scholar 

  28. Klein, M., Eslami-Mossallam, B., Arroyo, D. G. & Depken, M. Hybridization kinetics explains CRISPR-Cas off-targeting rules. Cell Rep. 22, 1413–1423 (2018).

    CAS  PubMed  Google Scholar 

  29. Dagdas, Y. S., Chen, J. S., Sternberg, S. H., Doudna, J. A. & Yildiz, A. A conformational checkpoint between DNA binding and cleavage by CRISPR-Cas9. Sci. Adv. 3, eaao0027 (2017).

    PubMed  PubMed Central  Google Scholar 

  30. Haeussler, M. et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biol. 17, 148 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Singh, D. et al. Mechanisms of improved specificity of engineered Cas9s revealed by single-molecule FRET analysis. Nat. Struct. Mol. Biol. 25, 347–354 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Rueda, F. O. et al. Mapping the sugar dependency for rational generation of a DNA-RNA hybrid-guided Cas9 endonuclease. Nat. Commun. 8, 1610 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Kleinstiver, B. P. et al. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490–495 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Doench, J. G. et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat. Biotechnol. 32, 1262–1267 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    PubMed  PubMed Central  Google Scholar 

  39. Heller, I. et al. STED nanoscopy combined with optical tweezers reveals protein dynamics on densely covered DNA. Nat. Methods 10, 910–916 (2013).

    CAS  PubMed  Google Scholar 

  40. Gross, P., Farge, G., Peterman, E. J. G. & Wuite, G. J. L. Combining optical tweezers, single-molecule fluorescence microscopy, and microfluidics for studies of DNA-protein interactions. Methods Enzymol. 475, 427–453 (2010).

    CAS  PubMed  Google Scholar 

  41. Brouwer, I. et al. Sliding sleeves of XRCC4–XLF bridge DNA and connect fragments of broken DNA. Nature 535, 566–569 (2016).

    CAS  PubMed  Google Scholar 

  42. Zhang, X. et al. Revealing the competition between peeled ssDNA, melting bubbles, and S-DNA during DNA overstretching by single-molecule calorimetry. Proc. Natl Acad. Sci. USA 110, 3865–3870 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L. & Corn, J. E. Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339–344 (2016).

    CAS  PubMed  Google Scholar 

  44. Modesti, M. Fluorescent labeling of proteins. Methods Mol. Biol. 783, 101–120 (2011).

    CAS  PubMed  Google Scholar 

  45. Krasikova, Y. S., Rechkunova, N. I., Maltseva, E. A., Petruseva, I. O. & Lavrik, O. I. Localization of xeroderma pigmentosum group A protein and replication protein A on damaged DNA in nucleotide excision repair. Nucleic Acids Res. 38, 8083–8094 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, R., Subramanyam, S., Elcock, A. H., Spies, M. & Wold, M. S. Dynamic binding of replication protein a is required for DNA repair. Nucleic Acids Res. 44, 5758–5772 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. King, G. A., Peterman, E. J. G. & Wuite, G. J. L. Unravelling the structural plasticity of stretched DNA under torsional constraint. Nat. Commun. 7, 11810 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. van Mameren, J. et al. Unraveling the structure of DNA during overstretching by using multicolor, single-molecule fluorescence imaging. Proc. Natl Acad. Sci. USA 106, 18231–18236 (2009).

    PubMed  PubMed Central  Google Scholar 

  49. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Eyquem, J. et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 543, 113–117 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Ren, J. et al. Multiplex genome editing to generate universal car T cells resistant to PD1 inhibition. Clin. Cancer Res. 23, 2255–2266 (2017).

    CAS  PubMed  Google Scholar 

  52. Gong, S., Yu, H. H., Johnson, K. A. & Taylor, D. W. DNA unwinding is the primary determinant of CRISPR-Cas9 activity. Cell Rep. 22, 359–371 (2018).

    CAS  PubMed  Google Scholar 

  53. O’Geen, H., Henry, I. M., Bhakta, M. S., Meckler, J. F. & Segal, D. J. A genome-wide analysis of Cas9 binding specificity using ChIP-seq and targeted sequence capture. Nucleic Acids Res. 43, 3389–3404 (2015).

    PubMed  PubMed Central  Google Scholar 

  54. Senavirathne, G. et al. Activation-induced deoxycytidine deaminase (AID) co-transcriptional scanning at single-molecule resolution. Nat. Commun. 6, 10209 (2015).

    CAS  PubMed  Google Scholar 

  55. Steger, C. An unbiased detector of curvilinear structures. IEEE. Trans. Pattern Anal. Mach. Intell. 20, 113–125 (1998).

    Google Scholar 

  56. Flores, O. & Orozco, M. nucleR: a package for non-parametric nucleosome positioning. Bioinformatics 27, 2149–2150 (2011).

    CAS  PubMed  Google Scholar 

  57. Bailey, T. L. et al. Meme Suite: tools for motif discovery and searching. Nucleic Acids Res. 37, W202–W208 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Lamichhane, R., Solem, A., Black, W. & Rueda, D. Single-molecule FRET of protein–nucleic acid and protein–protein complexes: surface passivation and immobilization. Methods 52, 192–200 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhao, R. & Rueda, D. RNA folding dynamics by single-molecule fluorescence resonance energy transfer. Methods 49, 112–117 (2009).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank E. Gordon (AstraZeneca) for preparing the Cas9 purified proteins, M. Modesti (CRCM, Marseille) for providing GFP-hRPA, and A. Candelli and J. Cabanas (LUMICKS B.V.) for assistance with the initial tweezers experiments. The Single Molecule Imaging Group is funded by a core grant of the MRC-London Institute of Medical Sciences (UKRI MC-A658-5TY10), a Wellcome Trust Collaborative Grant (206292/Z/17/Z), a Leverhulme Grant (RPG-2016-214), and a BBSRC CASE-studentship (to M.D.N.). The Computational Regulatory Genomics Group is supported by the Medical Research Council UK (MC UP 1102/1), L.R. is supported by the Wellcome Trust (106954) and N.C. is supported by EMBO LTF (EMBO ALTF 1279-2016).

Author information

Authors and Affiliations

Authors

Contributions

M.D.N., B.J.T., M.E.C. and D.S.R. designed the studies. M.D.N. and R.P.C.D. performed the tweezers experiments. L.R., N.C. and B.L. carried out the de novo motif search. M.D.N. and S.A. did the smFRET and cleavage experiments. M.D.N. and D.S.R. analyzed the data and wrote the manuscript with input from all the authors.

Corresponding authors

Correspondence to Maria Emanuela Cuomo or David S. Rueda.

Ethics declarations

Competing interests

R.P.C.D. is an employee and shareholder of LUMICKS.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 No binding is observed with crRNA alone, with crRNA:tracrRNA or crRNA:Cas9 at low or high force or with nt-crRNA:tracrRNA:dCas9 at low force.

a Kymograph of force-stretched λ-DNA in the presence of 5′-Cy3-λ2-crRNA without tracrRNA or dCas9. b Kymograph of force-stretched λ-DNA in the presence of annealed 5′-Cy3-λ2-crRNA:tracrRNA. c Kymograph of force-stretched λ-DNA in the presence of dCas9 and 5’-Cy3-λ2-crRNA without tracrRNA. d Kymograph of low force-stretched (10 pN) λ-DNA in the presence of dCas9 and 5′-Cy3-nt-crRNA:tracrRNA. Vertical scale bar = 1 μm. e Bulk cleavage assay of Cy3-labeled λ2 On-Target DNA (20 nM) (Supplementary Table 1) with λ2-crRNA:tracrRNA:wtCas9 (50 nM). Visualized by 18% denaturing PAGE with separate fluorescent detection for Cy3 and Cy5 fluorescence. Lane 1, no Cas9 complex, Lane 2, unlabeled λ2-crRNA:tracrRNA and wtCas9 (Supplementary Table 1), Lane 3 5′-Cy5-λ2-crRNA:tracrRNA and wtCas9 (Supplementary Table 1). Cy5 labeling of the modified crRNA does not affect λ2 target cleavage efficiency. f Image Analysis Pipeline i) Kymographs are time-binned and their average intensity calculated ii) Bead edge is defined as point where intensity hits background intensity of region known to be DNA iii) Bead trimmed DNA is linearly mapped across known λ-DNA sequence, alignments corrected on the basis of on-target, then smoothed by FFT and intensities within each time window normalized iv) Binding events are detected using ridge detection ImageJ plugin v) Lengths of binding events are calculated and used to build dwell-time histograms. Vi) Position of each event is calculated, mapped back to λ-DNA sequence and used to build histograms of binding sites.

Supplementary Figure 2 Increased force stabilizes the long lived off-target binding mode.

a Dwell-time histogram of Cy3-λ2-crRNA:tracrRNA:dCas9 off-target λ-DNA binding events induced at forces from 20–50 pN. Bin size = 2 s. b Integrated cumulative probability density plots with corresponding exponential fits and fitting values. Data at 20 pN was fit with single exponential. Data for 30–50 pN was fit with a double exponential. Fit coefficient ± s.d.

Supplementary Figure 3 Off-target binding locations occur at non-random, guide sequence-specific sites.

a GC content or PAM site distribution across the λ-genome. Distribution of canonical PAM sites (NGG) and alternative PAM sites (NAG) across λ-DNA (Top). Distribution of GC content across λ-DNA (Bottom). Bin size = 200 bp. b Average time-binned intensity histograms for λ2, λ4 and nt guide. Off-targets are induced on force stretching DNA to 40 pN and show recurrent non-random off-target binding locations. Arrows marks on-target sites for λ2 (18.5 kb) and λ4 (30.5 kb). c,d,e Histograms (500 nt bin-width) of mapped off-target binding events at 40 pN for 3 guides (λ2-, λ4- and n.t.-crRNA, respectively). Each guide experiment was repeated in triplicate (λ2-crRNA, n = 278, 645 and 311; λ4-crRNA, n = 437, 210 and 150; n.t.-crRNA, n = 605, 876 and 469). f Pearson’s correlation analysis between count normalized histograms of each binding localization repeat (Rep. 1–3) and guide. A strong correlation (dark red) is observed between experiments with the same crRNA guide but not (light red) between different guides, demonstrating that force-induced off-target binding is DNA and crRNA sequence specific. g Kymograph of λ-DNA in the presence of dCas9 with 5′-Cy3-nt-crRNA:tracrRNA (green) and hRPA-eGFP (blue). Off-target binding is observed with increasing force. hRPA binding is observed on nicked DNA and large ssDNA regions. To maintain constant force, extension increases to compensate for DNA nick formation. Off-target Cas9 binding and hRPA binding are mutually exclusive suggesting that two DNA strands are required for Cas9 binding. h Kymograph of 100 nM wtCas9 in complex with non-target 5′-Cy3-nt-crRNA:tracrRNA. Overstretched nicked DNA generates large ssDNA regions, which Cas9 no longer binds. Source data.

Source data

Supplementary Figure 4 Sequence analysis of Cas9 off-target binding.

a Example processing of the λ2-crRNA intensity. Raw intensities were normalized to maximum signal, aligned to account for small peak shifts caused by force stretching the λ-DNA, and smoothed using FFT function to reduce the noise. b Heatmap representation of guide-RNA sequence weighted nucleotide matches that fall within a peak, that is Cas9 binding event (Type of match: yes) versus outside the peak, that is Cas9 not bound (Type of match: no). A minimum 60% sequence match (12/20 nucleotides) of crRNA is defined. Identified matches were extracted and its sequence composition analyzed. All matching sequences were ordered by distance to the closest peak maxima and clustered according to in vs outside peak location (in peak is up to 200 bp distance from the peak maxima). Matching to the PAM sequence (NGG) was analyzed separately.

Supplementary Figure 5 Example FRET trajectories for binding to bubble constructs and FRET histograms of crRNA only, double-stranded PAM and ssDNA.

a FRET histograms of the full bubble DNA constructs with 10 nM 5′-Cy5-λ2-crRNA:tracrRNA only (no dCas9). A small population (21%, green, n = 1530) of high FRET molecules bind on-target bubble DNA but not off-targets 1–3 (n = 1610, 2094, and 2120 respectively). b Representative FRET trajectories for λ2-crRNA:tracrRNA:dCas9 binding on-target and off-target bubble constructs. Stable high FRET is observed for on-target and off-targets 1 and 3 followed by single-step photobleaching. For off-target 2 dynamic equilibrium between FRET of ~0.55 and ~0.85 is observed followed by single-step photobleaching. c FRET histograms for ssDNA targets with 10 nM 5′-Cy5-λ2-crRNA:tracrRNA:dCas9. A small population (25%, green, n = 3717) of high FRET molecules bind the on-target constructs but not off-targets 1–3 (n = 2234, n = 1499, n = 1012 for OT1, OT2 and OT3 respectively). d FRET histograms for double-stranded PAM constructs with 10 nM 5′-Cy5-λ2-crRNA:tracrRNA:dCas9 show a small high FRET population (20%, n = 1976; 22%, n = 3062; 23%, n = 2127; 31%, n = 2874; for ON, OT1, OT2 and OT3, respectively). e FRET histograms for a small (6 nt) PAM-proximal bubble. A bound population is observed for on-target (FRET = 0.97 ± 0.02, 36%, n = 3863), OT1 (FRET = 0.23 ± 0.02, 52%, n = 2466) and OT2 (FRET = 0.27 ± 0.02, 75%, n = 3704) but not for OT3 (n = 626).

Supplementary Figure 6 Bulk cleavage assays with various bubble sizes and location, and bona fide cellular EMX1-1 off-targets.

a Cleavage assay for OT2 with different PAM sequences. Efficient cleavage is only observed in the presence of canonical (NGG) or non-canonical (NAG) PAM sequences with equal efficiency. b Cleavage assay for OT3 with λ2-crRNA:tracrRNA:wtCas9, and bubbles from 0–20 nt. Cleavage is only observed with ≥14 nt bubbles. c Cleavage assay for ON and OT1–3 with small (6 nt) PAM-proximal, middle and -distal bubbles. Cleavage is only observed for ON. d Cleavage assay for ON and OT1–3 with double-stranded (ds), single-stranded (ss), double-stranded PAM (dsPAM) and full bubble constructs (bub). Most efficient OT cleavage is only observed in the presence both dsPAM and non-target strand. All with 100 nM wtCas9 complex (see methods). e Genomic locations and sequences of EMX1-1 target (ON) and four off-targets (OT) identified with CIRCLE-seq and validated in cells. Seed sequence in bold, mismatches (4 each) highlighted in red and underlined. f Denaturing PAGE cleavage assay (left) and quantification (right, n = 3) shows that CRISPR-Cas9 cuts bubbled (bub) OT substrates with four mismatches and not double-stranded ones (ds). Error bars = s.d.

Supplementary information

Supplementary Figures, Tables and Notes

Supplementary Figures 1–6, Supplementary Tables 1–3 and Supplementary Note 1

Reporting Summary

Supplementary Video 1

DNA stretching induces Cas9 off-target binding Force stretching λ-DNA from 5 pN to 50 pN induces reversible off-target binding at multiple sites. Confocal time-lapse movie initially shows a single λ2-crRNA-dCas9 complex (green dot) bound on-target to λ-DNA held between two beads by optical tweezers at low force (5 pN). As force is slowly increased to 50 pN, multiple off-target binding events appear. Off-targets dissociate while the force is decreased back to 5 pN, and only the on-target complex remains bound. Time stamp in mm:ss.

Supplementary Dataset 1

Gel images for Fig. 4f. Raw images of gels used for quantification for Figure 4f.

Source data for Fig. 2b,c

Table of dwell times at different forces of all events used for kinetic analysis in Figure 2b,c and Supplementary Figure 2.

Source data for Fig. 3c

Table of locations of detected binding events used to generate binding distribution histograms in Figure 3c and Supplementary Figure 3c–f. Contains three repeats of each of the three guides tested.

Source data for Fig. 4f

Quantifications of cleavage efficiencies of different bubble sizes for the three off-targets tested used to plot Figure 4f.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Newton, M.D., Taylor, B.J., Driessen, R.P.C. et al. DNA stretching induces Cas9 off-target activity. Nat Struct Mol Biol 26, 185–192 (2019). https://doi.org/10.1038/s41594-019-0188-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0188-z

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