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.

  • Review Article
  • Published:

Tools for experimental and computational analyses of off-target editing by programmable nucleases

Abstract

Genome editing using programmable nucleases is revolutionizing life science and medicine. Off-target editing by these nucleases remains a considerable concern, especially in therapeutic applications. Here we review tools developed for identifying potential off-target editing sites and compare the ability of these tools to properly analyze off-target effects. Recent advances in both in silico and experimental tools for off-target analysis have generated remarkably concordant results for sites with high off-target editing activity. However, no single tool is able to accurately predict low-frequency off-target editing, presenting a bottleneck in therapeutic genome editing, because even a small number of cells with off-target editing can be detrimental. Therefore, we recommend that at least one in silico tool and one experimental tool should be used together to identify potential off-target sites, and amplicon-based next-generation sequencing (NGS) should be used as the gold standard assay for assessing the true off-target effects at these candidate sites. Future work to improve off-target analysis includes expanding the true off-target editing dataset to evaluate new experimental techniques and to train machine learning algorithms; performing analysis using the particular genome of the cells in question rather than the reference genome; and applying novel NGS techniques to improve the sensitivity of amplicon-based off-target editing quantification.

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: Schematics showing four major classes of programmable nucleases.
Fig. 2: Schematics showing in vitro and in vivo experimental techniques most commonly used to characterize off-target cutting by CRISPR–Cas9.
Fig. 3: The ability of various off-target site identification algorithms to correctly rank experimentally confirmed true off-target sites.
Fig. 4: Recommended workflow for identifying CRISPR–Cas9 off-target editing.

Similar content being viewed by others

Data availability

The data sources are available at https://github.com/baolab-rice/OT-review. Supplementary Tables 3, 4 and 5 contain the data used in the computational techniques performance assessment.

Code availability

The processing scripts are available at https://github.com/baolab-rice/OT-review.

References

  1. Kim, Y. G., Cha, J. & Chandrasegaran, S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc. Natl Acad. Sci. USA 93, 1156–1160 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Porteus, M. H. & Baltimore, D. Chimeric nucleases stimulate gene targeting in human cells. Science 300, 763 (2003).

    Article  PubMed  Google Scholar 

  3. Urnov, F. D. et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature 435, 646–651 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, 7879–7879 (2011).

    Article  CAS  PubMed Central  Google Scholar 

  5. Miller, J. C. et al. A TALE nuclease architecture for efficient genome editing. Nat. Biotechnol. 29, 143–148 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Mussolino, C. et al. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 39, 9283–9293 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. 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 

  9. Cobb, R. E., Wang, Y. J. & Zhao, H. M. High-efficiency multiplex genome editing of streptomyces species using an engineered CRISPR/Cas system. ACS Synth. Biol. 4, 723–728 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Gaudelli, N. M. et al. Programmable base editing of A.T to G.C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Carroll, D. Genome engineering with zinc-finger nucleases. Genetics 188, 773–782 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Joung, J. K. & Sander, J. D. TALENs: a widely applicable technology for targeted genome editing. Nat. Rev. Mol. Cell Biol. 14, 49–55 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Cox, D. B., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121–131 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wagner, D. L. et al. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat. Med. 25, 242–248 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Charlesworth, C. T. et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nat. Med. 25, 249–254 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Simhadri, V. L. et al. Prevalence of pre-existing antibodies to CRISPR-associated nuclease Cas9 in the USA population. Mol. Ther. Methods Clin. Dev. 10, 105–112 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, A. et al. AAV-CRISPR gene editing is negated by pre-existing immunity to Cas9. Mol. Ther. 28, 1432–1441 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tong, S., Moyo, B., Lee, C. M., K., L. & Bao, G. Engineered materials for in vivo delivery of genome-editing machinery. Nat. Rev. Mater. 4, 726–737 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. 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 

  25. Cradick, T. J., Fine, E. J., Antico, C. J. & Bao, G. CRISPR/Cas9 systems targeting beta-globin and CCR5 genes have substantial off-target activity. Nucleic Acids Res. 41, 9584–9592 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. 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).

  28. Qiu, P. et al. Mutation detection using Surveyor nuclease. Biotechniques 36, 702–707 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Kim, H. J., Lee, H. J., Kim, H., Cho, S. W. & Kim, J. S. Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. 19, 1279–1288 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Brinkman, E. K., Chen, T., Amendola, M. & van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res. 42, e168 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Hsiau, T. et al. Inference of CRISPR edits from Sanger trace data. Preprint at https://www.biorxiv.org/content/10.1101/251082v3 (2019).

  32. Potapov, V. & Ong, J. L. Examining sources of error in PCR by single-molecule sequencing. PLoS ONE 12, e0169774 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Jarrett, K. E. et al. Somatic genome editing with CRISPR/Cas9 generates and corrects a metabolic disease. Sci. Rep. 7, 44624 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zykovich, A., Korf, I. & Segal, D. J. Bind-n-Seq: high-throughput analysis of in vitro protein–DNA interactions using massively parallel sequencing. Nucleic Acids Res. 37, e151 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat. Biotechnol. 26, 808–816 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Segal, D. J. et al. Evaluation of a modular strategy for the construction of novel polydactyl zinc finger DNA-binding proteins. Biochemistry 42, 2137–2148 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Gupta, A., Meng, X., Zhu, L. J., Lawson, N. D. & Wolfe, S. A. Zinc finger protein-dependent and -independent contributions to the in vivo off-target activity of zinc finger nucleases. Nucleic Acids Res. 39, 381–392 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Segal, D. J., Dreier, B., Beerli, R. R. & Barbas, C. F. 3rd Toward controlling gene expression at will: selection and design of zinc finger domains recognizing each of the 5′-GNN-3′ DNA target sequences. Proc. Natl Acad. Sci. USA 96, 2758–2763 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bulyk, M. L., Huang, X., Choo, Y. & Church, G. M. Exploring the DNA-binding specificities of zinc fingers with DNA microarrays. Proc. Natl Acad. Sci. USA 98, 7158–7163 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zhang, L. et al. Systematic in vitro profiling of off-target affinity, cleavage and efficiency for CRISPR enzymes. Nucleic Acids Res. 48, 5037–5053 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Fu, B. X., 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 

  46. Huston, N. C. et al. Identification of guide-intrinsic determinants of Cas9 specificity. CRISPR J. 2, 172–185 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. 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  PubMed  PubMed Central  Google Scholar 

  50. Lazzarotto, C. R. et al. CHANGE-seq reveals genetic and epigenetic effects on CRISPR–Cas9 genome-wide activity. Nat Biotechnol. 38, 1317–1327 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kim, D. & Kim, J. S. DIG-seq: a genome-wide CRISPR off-target profiling method using chromatin DNA. Genome Res. 28, 1894–1900 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. 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 

  53. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–191 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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 

  57. Gabriel, R. et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity. Nat. Biotechnol. 29, 816–823 (2011).

    Article  CAS  PubMed  Google Scholar 

  58. Hu, J. et al. Detecting DNA double-stranded breaks in mammalian genomes by linear amplification-mediated high-throughput genome-wide translocation sequencing. Nat. Protoc. 11, 853–871 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hruscha, A. et al. Efficient CRISPR/Cas9 genome editing with low off-target effects in zebrafish. Development 140, 4982–4987 (2013).

    Article  CAS  PubMed  Google Scholar 

  60. Zhu, X. et al. An efficient genotyping method for genome-modified animals and human cells generated with CRISPR/Cas9 system. Sci. Rep. 4, 6420 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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).

    PubMed  PubMed Central  Google Scholar 

  62. Montague, T. G., Cruz, J. M., Gagnon, J. A., Church, G. M. & Valen, E. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing. Nucleic Acids Res. 42, W401–W407 (2014).

  63. O’Brien, A. & Bailey, T. L. GT-Scan: identifying unique genomic targets. Bioinformatics 30, 2673–2675 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Langdon, W. B. Performance of genetic programming optimised Bowtie2 on genome comparison and analytic testing (GCAT) benchmarks. BioData Min. 8, 1 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Heigwer, F., Kerr, G. & Boutros, M. E-CRISP: fast CRISPR target site identification. Nat. Methods 11, 122–123 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. Moreno-Mateos, M. A. et al. CRISPRscan: designing highly efficient sgRNAs for CRISPR–Cas9 targeting in vivo. Nat. Methods 12, 982–988 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Cancellieri, S., Canver, M. C., Bombieri, N., Giugno, R. & Pinello, L. CRISPRitz: rapid, high-throughput, and variant-aware in silico off-target site identification for CRISPR genome editing. Bioinformatics 36, 2001–2008 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  68. Zschemisch, N. H. et al. Zinc-finger nuclease mediated disruption of Rag1 in the LEW/Ztm rat. BMC Immunol. 13, 60 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Watanabe, T. et al. Non-transgenic genome modifications in a hemimetabolous insect using zinc-finger and TAL effector nucleases. Nat. Commun. 3, 1017 (2012).

    Article  PubMed  CAS  Google Scholar 

  70. Sebastiano, V. et al. In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem Cells 29, 1717–1726 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Huang, P. et al. Heritable gene targeting in zebrafish using customized TALENs. Nat. Biotechnol. 29, 699–700 (2011).

    Article  PubMed  CAS  Google Scholar 

  72. Lei, Y. et al. Efficient targeted gene disruption in Xenopus embryos using engineered transcription activator-like effector nucleases (TALENs). Proc. Natl Acad. Sci. USA 109, 17484–17489 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Heigwer, F. et al. E-TALEN: a web tool to design TALENs for genome engineering. Nucleic Acids Res. 41, e190 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kim, Y. et al. A library of TAL effector nucleases spanning the human genome. Nat. Biotechnol. 31, 251–258 (2013).

    Article  CAS  PubMed  Google Scholar 

  75. Doyle, E. L. et al. TAL effector-nucleotide targeter (TALE-NT) 2.0: tools for TAL effector design and target prediction. Nucleic Acids Res. 40, W117–W122 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Grau, J., Boch, J. & Posch, S. TALENoffer: genome-wide TALEN off-target prediction. Bioinformatics 29, 2931–2932 (2013).

    Article  CAS  PubMed  Google Scholar 

  77. Streubel, J., Blücher, C., Landgraf, A. & Boch, J. TAL effector RVD specificities and efficiencies. Nat. Biotechnol. 30, 593–595 (2012).

    Article  CAS  PubMed  Google Scholar 

  78. Fine, E. J., Cradick, T. J., Zhao, C. L., Lin, Y. & Bao, G. An online bioinformatics tool predicts zinc finger and TALE nuclease off-target cleavage. Nucleic Acids Res. 42, e42 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Meckler, J. F. et al. Quantitative analysis of TALE–DNA interactions suggests polarity effects. Nucleic Acids Res. 41, 4118–4128 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. 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  PubMed  PubMed Central  CAS  Google Scholar 

  81. 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 

  82. Jacquin, A. L. S., Odom, D. T. & Lukk, M. Crisflash: open-source software to generate CRISPR guide RNAs against genomes annotated with individual variation. Bioinformatics 35, 3146–3147 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Xiao, A. et al. CasOT: a genome-wide Cas9/gRNA off-target searching tool. Bioinformatics 30, 1180–1182 (2014).

    Article  CAS  PubMed  Google Scholar 

  84. 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  CAS  Google Scholar 

  85. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Singh, R., Kuscu, C., Quinlan, A., Qi, Y. & Adli, M. Cas9-chromatin binding information enables more accurate CRISPR off-target prediction. Nucleic Acids Res. 43, e118 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Thurman, R. E. et al. The accessible chromatin landscape of the human genome. Nature 489, 75–82 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Abadi, S., Yan, W. X., Amar, D. & Mayrose, I. A machine learning approach for predicting CRISPR–Cas9 cleavage efficiencies and patterns underlying its mechanism of action. PLoS Comput. Biol. 13, e1005807 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Peng, H., Zheng, Y., Zhao, Z., Liu, T. & Li, J. Recognition of CRISPR/Cas9 off-target sites through ensemble learning of uneven mismatch distributions. Bioinformatics 34, i757–i765 (2018).

    Article  CAS  PubMed  Google Scholar 

  91. Listgarten, J. et al. Prediction of off-target activities for the end-to-end design of CRISPR guide RNAs. Nat. Biomed. Eng. 2, 38–47 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lin, J. & Wong, K. C. Off-target predictions in CRISPR–Cas9 gene editing using deep learning. Bioinformatics 34, i656–i663 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Chuai, G. et al. DeepCRISPR: optimized CRISPR guide RNA design by deep learning. Genome Biol. 19, 80 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Zhang, S. X., Li, X. T., Lin, Q. Z. & Wong, K. C. Synergizing CRISPR/Cas9 off-target predictions for ensemble insights and practical applications. Bioinformatics 35, 1108–1115 (2019).

    Article  CAS  PubMed  Google Scholar 

  95. Luo, J. S., Chen, W., Xue, L. & Tang, B. Prediction of activity and specificity of CRISPR-Cpf1 using convolutional deep learning neural networks. BMC Bioinformatics 20, 332 (2019).

  96. Kim, D. et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat. Biotechnol. 35, 797–797 (2017).

    Article  CAS  PubMed  Google Scholar 

  97. Rees, H. A. & Liu, D. R. Publisher Correction: Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 801 (2018).

    Article  CAS  PubMed  Google Scholar 

  98. Zuo, E. W. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289–292 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jin, S. et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364, 292–295 (2019).

    Article  CAS  PubMed  Google Scholar 

  100. Grunewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 5, eaax5717 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  102. Zhou, C. Y. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275–278 (2019).

    Article  CAS  PubMed  Google Scholar 

  103. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, aaf8729 (2016).

    Article  PubMed  CAS  Google Scholar 

  105. Kim, D., Kim, D. E., Lee, G., Cho, S. I. & Kim, J. S. Genome-wide target specificity of CRISPR RNA-guided adenine base editors. Nat. Biotechnol. 37, 430–435 (2019).

    Article  CAS  PubMed  Google Scholar 

  106. Liang, P. P. et al. Genome-wide profiling of adenine base editor specificity by EndoV-seq. Nat. Commun. 10, 67 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Yu, Y. et al. Cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity. Nat. Commun. 11, 2052 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Doman, J. L., Raguram, A., Newby, G. A. & Liu, D. R. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat. Biotechnol. 38, 620–628 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  110. Kim, D., Luk, K., Wolfe, S. A. & Kim, J. S. Evaluating and enhancing target specificity of gene-editing nucleases and deaminases. Annu. Rev. Biochem. 88, 191–220 (2019).

    Article  CAS  PubMed  Google Scholar 

  111. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224–226 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. 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  PubMed  PubMed Central  Google Scholar 

  113. 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 

  114. Park, S. H. et al. Highly efficient editing of the beta-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease. Nucleic Acids Res. 47, 7955–7972 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Gomez-Ospina, N. et al. Human genome-edited hematopoietic stem cells phenotypically correct Mucopolysaccharidosis type I. Nat. Commun. 10, 4045 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Pavel-Dinu, M. et al. Gene correction for SCID-X1 in long-term hematopoietic stem cells. Nat. Commun. 10, 1634 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Vaidyanathan, S. et al. High-efficiency, selection-free gene repair in airway stem cells from cystic fibrosis patients rescues CFTR function in differentiated epithelia. Cell Stem Cell 26, 161–171 e164 (2020).

    Article  CAS  PubMed  Google Scholar 

  118. Pedregosa, E. et al. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 12, 2825–2830 (2011).

    Google Scholar 

  119. Sledzinski, P., Nowaczyk, M. & Olejniczak, M. Computational tools and resources supporting CRISPR–Cas experiments. Cells 9, 1288 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  120. Park, J., Lim, K., Kim, J. S. & Bae, S. Cas-analyzer: an online tool for assessing genome editing results using NGS data. Bioinformatics 33, 286–288 (2017).

    Article  CAS  PubMed  Google Scholar 

  121. Guell, M., Yang, L. & Church, G. M. Genome editing assessment using CRISPR Genome Analyzer (CRISPR-GA). Bioinformatics 30, 2968–2970 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. DiGiusto, D. L. et al. Preclinical development and qualification of ZFN-mediated CCR5 disruption in human hematopoietic stem/progenitor cells. Mol. Ther. Methods Clin. Dev. 3, 16067 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Fraietta, J. A. et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature 558, 307–312 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hacein-Bey-Abina, S. et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Miller, J. C. et al. Enhancing gene editing specificity by attenuating DNA cleavage kinetics. Nat. Biotechnol. 37, 945–952 (2019).

    Article  CAS  PubMed  Google Scholar 

  126. Schneider, V. A. et al. Evaluation of GRCh38 and de novo haploid genome assemblies demonstrates the enduring quality of the reference assembly. Genome Res. 27, 849–864 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Sherman, R. M. et al. Assembly of a pan-genome from deep sequencing of 910 humans of African descent. Nat. Genet. 51, 30–35 (2019).

    Article  CAS  PubMed  Google Scholar 

  128. Cradick, T. J., Ambrosini, G., Iseli, C., Bucher, P. & McCaffrey, A. P. ZFN-site searches genomes for zinc finger nuclease target sites and off-target sites. BMC Bioinformatics 12, 152 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Iseli, C., Ambrosini, G., Bucher, P. & Jongeneel, C. V. Indexing strategies for rapid searches of short words in genome sequences. PLoS ONE 2, e579 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Mandell, J. G. & Barbas, C. F. 3rd Zinc finger tools: custom DNA-binding domains for transcription factors and nucleases. Nucleic Acids Res. 34, W516–W523 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Altschul, S. F., Gish, W., Miller, W., Myers, E. W. & Lipman, D. J. Basic local alignment search tool. J. Mol. Biol. 215, 403–410 (1990).

    Article  CAS  PubMed  Google Scholar 

  132. Liu, G., Zhang, Y. & Zhang, T. Computational approaches for effective CRISPR guide RNA design and evaluation. Comput. Struct. Biotechnol. J. 18, 35–44 (2020).

    Article  CAS  PubMed  Google Scholar 

  133. McKenna, A. & Shendure, J. FlashFry: a fast and flexible tool for large-scale CRISPR target design. BMC Biol. 16, 74 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Institutes of Health (UG3HL151545, R01HL152314 and OT2HL154977 to G.B.) and the Cancer Prevention and Research Institute of Texas (RR140081 to G.B.).

Author information

Authors and Affiliations

Authors

Contributions

G.B. and X.R.B. designed the study. X.R.B. and C.M.L. performed the evaluation of experimental methods. Y.P. and X.R.B. performed the evaluation of computational methods. All authors contributed to the discussion and writing of the manuscript.

Corresponding author

Correspondence to Gang Bao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks the anonymous reviewers 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2.

Supplementary Table 1

Structures and training sets of machine learning-based algorithms for off-target prediction.

Supplementary Table 2

List of datasets from the Sequence Read Archive used to calculate on-target enrichment for performance comparison of experimental techniques. All genomic coordinates reference the hg38 human genome assembly.

Supplementary Table 3

Components of the true-positive list used in the analysis for performance comparison of computational techniques, taken from nine studies that used amplicon-specific experimental techniques to detect off-target editing rates. ‘OT’ in this table stands for off-target.

Supplementary Table 4

The full off-target dataset used in the performance assessment of computational techniques. Potential off-target sites of 27 gRNAs from nine studies shown in Supplementary Table 3 were screened by Cas-OFFinder, allowing up to four mismatches and one base DNA/RNA bulge and scored by each of the algorithms. ‘noind’: DNA/RNA sequences before alignments. Note that, after introducing bulges, one locus might be called by Cas-OFFinder multiple times with different alignment patterns, leading to different scores. These sites were treated the same as other off-target sites without manipulation. Source data for Supplementary Figure 1 and Fig. 3a.

Supplementary Table 5

The off-target dataset of novel gRNAs used in the performance assessment of computational techniques. Potential off-target sites and scores of four gRNAs that were not in the CRISPOR dataset1. These were screened by Cas-OFFinder, allowing up to four mismatches and one base DNA/RNA bulge and scored by each of the algorithms. Note that, after introducing bulges, one locus might be called by Cas-OFFinder multiple times with different alignment patterns, leading to different scores. These sites were treated the same as other off-target sites without manipulation. ‘noind’: DNA/RNA sequences before alignments. Source data for Supplementary Fig. 2 and Fig. 3b.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bao, X.R., Pan, Y., Lee, C.M. et al. Tools for experimental and computational analyses of off-target editing by programmable nucleases. Nat Protoc 16, 10–26 (2021). https://doi.org/10.1038/s41596-020-00431-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-020-00431-y

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