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.

  • Protocol
  • Published:

CRISPR off-target detection with DISCOVER-seq

Abstract

DISCOVER-seq (discovery of in situ Cas off-targets and verification by sequencing) is a broadly applicable approach for unbiased CRISPR–Cas off-target identification in cells and tissues. It leverages the recruitment of DNA repair factors to double-strand breaks (DSBs) after genome editing with CRISPR nucleases. Here, we describe a detailed experimental protocol and analysis pipeline with which to perform DISCOVER-seq. The principle of this method is to track the precise recruitment of MRE11 to DSBs by chromatin immunoprecipitation followed by next-generation sequencing. A customized open-source bioinformatics pipeline, BLENDER (blunt end finder), then identifies off-target sequences genome wide. DISCOVER-seq is capable of finding and measuring off-targets in primary cells and in situ. The two main advantages of DISCOVER-seq are (i) low false-positive rates because DNA repair enzyme binding is required for genome edits to occur and (ii) its applicability to a wide variety of systems, including patient-derived cells and animal models. The whole protocol, including the analysis, can be completed within 2 weeks.

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: Overview of DISCOVER-seq workflow and quality controls.
Fig. 2: Comparison of MRE11 antibodies.
Fig. 3: Quality controls of a DISCOVER-seq experiment.
Fig. 4: Example output of a successful DISCOVER-seq experiment.

Similar content being viewed by others

Data availability

Data that support the findings of this study have been deposited in SRA BioProject accession no. PRJNA509652.

Code availability

Our open-source analysis pipeline, called BLENDER (https://github.com/staciawyman/blender), is freely available under the GNU Affero General Public License. The code in this paper has been peer reviewed.

References

  1. Cheng, Y. & Tsai, S. Q. Illuminating the genome-wide activity of genome editors for safe and effective therapeutics. Genome Biol. 19, 226 (2018).

    Article  CAS  Google Scholar 

  2. Carroll, D. Collateral damage: benchmarking off-target effects in genome editing. Genome Biol. 20, 114 (2019).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Slaymaker, I. M. et al. Rationally engineered Cas9 nucleases with improved specificity. Science 351, 84–88 (2016).

    Article  CAS  Google Scholar 

  5. Vakulskas, C. A. et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat. Med 24, 1216–1224 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  7. Wienert, B. et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-seq. Science 364, 286–289 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Stracker, T. H. & Petrini, J. H. J. The MRE11 complex: starting from the ends. Nat. Rev. Mol. Cell Biol. 12, 90–103 (2011).

    Article  CAS  Google Scholar 

  9. Williams, R. S., Williams, J. S. & Tainer, J. A. Mre11-Rad50-Nbs1 is a keystone complex connecting DNA repair machinery, double-strand break signaling, and the chromatin template. Biochem. Cell Biol. 85, 509–520 (2007).

    Article  CAS  Google Scholar 

  10. Kobayashi, J. Molecular mechanism of the recruitment of NBS1/hMRE11/hRAD50 complex to DNA double-strand breaks: NBS1 binds to gamma-H2AX through FHA/BRCT domain. J. Radiat. Res. 45, 473–478 (2004).

    Article  CAS  Google Scholar 

  11. Richardson, C. D. et al. CRISPR-Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nat. Genet. 50, 1132–1139 (2018).

    Article  CAS  Google Scholar 

  12. 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).

    Article  Google Scholar 

  13. Lazzarotto, C. R. et al. Defining CRISPR-Cas9 genome-wide nuclease activities with CIRCLE-seq. Nat. Protoc. 13, 2615–2642 (2018).

    Article  CAS  Google Scholar 

  14. 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).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  17. 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  Google Scholar 

  18. Giannoukos, G. et al. UDiTaSTM, a genome editing detection method for indels and genome rearrangements. BMC Genomics 19, 212 (2018).

    Article  Google Scholar 

  19. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Yan, W. X. et al. BLISS is a versatile and quantitative method for genome-wide profiling of DNA double-strand breaks. Nat. Commun. 8, 15058 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  23. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    Article  CAS  Google Scholar 

  24. Rass, E. et al. Role of Mre11 in chromosomal nonhomologous end joining in mammalian cells. Nat. Struct. Mol. Biol. 16, 819–824 (2009).

    Article  CAS  Google Scholar 

  25. Xie, A., Kwok, A. & Scully, R. Role of mammalian Mre11 in classical and alternative nonhomologous end joining. Nat. Struct. Mol. Biol. 16, 814–818 (2009).

    Article  CAS  Google Scholar 

  26. Dinkelmann, M. et al. Multiple functions of MRN in end-joining pathways during isotype class switching. Nat. Struct. Mol. Biol. 16, 808–813 (2009).

    Article  CAS  Google Scholar 

  27. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  28. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE Blacklist: identification of problematic regions of the genome. Sci. Rep. 9, 9354 (2019).

    Article  Google Scholar 

  29. Wienert, B., Shin, J., Zelin, E., Pestal, K. & Corn, J. E. In vitro-transcribed guide RNAs trigger an innate immune response via the RIG-I pathway. PLoS Biol. 16, e2005840 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

J.E.C. and C.D.Y. were supported by the NOMIS Foundation, the Lotte and Adolf Hotz-Sprenger Stiftung, the Swiss National Science Foundation (310030_188858), the Fanconi Anemia Research Foundation, and the Bill and Melinda Gates Foundation. B.W. was supported by an NHMRC Early Career Fellowship. B.R.C and B.W. received support from the Gladstone Institutes and NIH grants R01-EY028249, R01-HL130533 and R01-HL13535801. S.K.W. was supported by the Li Ka Shing Foundation. The authors used the Vincent J. Coates Genomics Sequencing Laboratory at UC Berkeley, which is supported by NIH Instrumentation grant S10 OD018174.

Author information

Authors and Affiliations

Authors

Contributions

B.W., S.K.W. and J.E.C. wrote the manuscript with input from all authors. B.W. and S.K.W. developed the original experimental protocol in the J.E.C. lab. C.D.Y. performed ChIP-qPCR time-course experiments. B.W. performed all other experiments. S.K.W. analyzed ChIP-seq data and developed BLENDER software. B.R.C. provided reagents and expertise.

Corresponding authors

Correspondence to Beeke Wienert or Jacob E. Corn.

Ethics declarations

Competing interests

J.E.C. is a cofounder of Spotlight Therapeutics. J.E.C. has received sponsored research support from AstraZeneca and Pfizer. B.R.C. is a founder of Tenaya Therapeutics.

Additional information

Peer review information Nature Protocols thanks Britta Bouwman, Nicola Crosetto 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.

Related link

Key references using this protocol

Wienert, B. et al. Science 364, 286–289 (2019): https://science.sciencemag.org/content/364/6437/286

Richardson, C. D. et al. Nat. Genet. 50, 1132–1139 (2018): https://www.nature.com/articles/s41588-018-0174-0

Integrated supplementary information

Supplementary Figure 1 Uncropped western blot images.

(a) Whole blot images corresponding to Fig. 2a. Cross-reactivity between human and mouse of five different commercially available MRE11 antibodies as determined by Western Blot. Whole cell extracts (30 μg) from human K562 cells and murine B16-F10 cells were used for the analysis. (b) Whole blot images corresponding to Fig. 2c. Increasing amounts of antibody lead to more MRE11 depletion from cell lysate, as determined by Western Blot. However, increasing pulldown efficiency with larger amounts of antibody is not directly reflected in the levels of eluted MRE11, possibly due to non-linearity of Western blots. FT – flow through, IP - immunoprecipitation.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wienert, B., Wyman, S.K., Yeh, C.D. et al. CRISPR off-target detection with DISCOVER-seq. Nat Protoc 15, 1775–1799 (2020). https://doi.org/10.1038/s41596-020-0309-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-0309-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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