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:

CHANGE-seq reveals genetic and epigenetic effects on CRISPR–Cas9 genome-wide activity

Abstract

Current methods can illuminate the genome-wide activity of CRISPR–Cas9 nucleases, but are not easily scalable to the throughput needed to fully understand the principles that govern Cas9 specificity. Here we describe ‘circularization for high-throughput analysis of nuclease genome-wide effects by sequencing’ (CHANGE-seq), a scalable, automatable tagmentation-based method for measuring the genome-wide activity of Cas9 in vitro. We applied CHANGE-seq to 110 single guide RNA targets across 13 therapeutically relevant loci in human primary T cells and identified 201,934 off-target sites, enabling the training of a machine learning model to predict off-target activity. Comparing matched genome-wide off-target, chromatin modification and accessibility, and transcriptional data, we found that cellular off-target activity was two to four times more likely to occur near active promoters, enhancers and transcribed regions. Finally, CHANGE-seq analysis of six targets across eight individual genomes revealed that human single-nucleotide variation had significant effects on activity at ~15.2% of off-target sites analyzed. CHANGE-seq is a simplified, sensitive and scalable approach to understanding the specificity of genome editors.

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

Fig. 1: Development and optimization of CHANGE-seq.
Fig. 2: High-throughput CHANGE-seq profiling of 110 therapeutic target sites reveals target site factors that affect Cas9 genome-wide specificity.
Fig. 3: Machine learning from large-scale CHANGE-seq datasets illuminates important predictors of off-target site activity.
Fig. 4: CHANGE-seq genome-wide activity profiles sensitively predict cellular specificity.
Fig. 5: Cas9 off-target activity is enriched in active chromatin states.
Fig. 6: CHANGE-seq detects impact of individual human genetic variation on Cas9 genome-wide activity.

Similar content being viewed by others

Data availability

High-throughput sequencing data generated during the study (CHANGE-seq, GUIDE-seq, ATAC-seq, histone modification ChIP–seq) are available from the NCBI Sequence Read Archive and Gene Expression Omnibus under accession numbers PRJNA625995 and GSE149295, respectively.

Code availability

Open-source code for analyzing CHANGE-seq or CIRCLE-seq (https://github.com/tsailabSJ/changeseq) and GUIDE-seq (https://github.com/aryeelab/guideseq), and for performing related machine learning analysis (https://github.com/ma-compbio/ChangeLocator), are available through GitHub.

References

  1. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Kocak, D. D. et al. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nat. Biotechnol. 37, 657–666 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Xie, K. & Yang, Y. RNA-guided genome editing in plants using a CRISPR–Cas system. Mol. Plant. 6, 1975–1983 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Cho, S. 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 

  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. 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 

  20. 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 

  21. 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 

  22. 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 

  23. 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 

  24. 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 

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

    Article  CAS  PubMed  Google Scholar 

  26. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Berg, D. E., Davies, J., Allet, B. & Rochaix, J. D. Transposition of R factor genes to bacteriophage lambda. Proc. Natl Acad. Sci. USA 72, 3628–3632 (1975).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Reznikoff, W. S. Transposon Tn5. Annu. Rev. Genet. 42, 269–286 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Adey, A. et al. Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition. Genome Biol. 11, R119 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Friedman, J. H. Greedy function approximation: a gradient boosting machine. Ann. Stat. 29, 1189–1232 (2001).

    Article  Google Scholar 

  32. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Yarrington, R. M., Verma, S., Schwartz, S., Trautman, J. K. & Carroll, D. Nucleosomes inhibit target cleavage by CRISPR-Cas9 in vivo. Proc. Natl Acad. Sci. USA 115, 9351–9358 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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 

  36. Scott, D. A. & Zhang, F. Implications of human genetic variation in CRISPR-based therapeutic genome editing. Nat. Med. 23, 1095–1101 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lessard, S. et al. Human genetic variation alters CRISPR-Cas9 on- and off-targeting specificity at therapeutically implicated loci. Proc. Natl Acad. Sci. USA 114, E11257–E11266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zook, J. M. et al. An open resource for accurately benchmarking small variant and reference calls. Nat. Biotechnol. 37, 561–566 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. 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 

  41. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Magoč, T. & Salzberg, S. L. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27, 2957–63 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–60 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Corces, R. M. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jiang, H., Lei, R., Ding, S.-W. & Zhu, S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics 15, 182 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  47. Cheng, Y. et al. Principles of regulatory information conservation between mouse and human. Nature 515, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Chao and R. Heath from the St. Jude Protein Production Core Facility for recombinant Tn5 production, G. Wu for variant calling, S. Olsen from the St. Jude Hartwell Center for whole-genome sequencing and RNA-seq, and J. Zook for discussions on Genome-in-a-Bottle data. Standard mapping and variant calling were performed by the Center for Applied Bioinformatics, a centralized shared resource, partly funded by NIH award no. P30CA021765. This work was supported by St. Jude Children’s Research Hospital and ALSAC, National Institutes of Health Common Fund Somatic Cell Genome Editing award no. U01EB029373 (to S.Q.T., Y.C., J.M., G.K. and S.M.), St. Jude Children’s Research Hospital Collaborative Research Consortium on Novel Gene Therapies for Sickle Cell Disease and the Doris Duke Charitable Foundation (grant no. 2017093), National Institute of General Medicine Science award no. R35GM133614 (to Y.C.) and National Heart, Lung, and Blood Institute award no. P01HL053749 (to S.Q.T.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. E.S. acknowledges support under the Cooperative Research Agreement between the University of Maryland and the National Institute of Standards and Technology Center for Nanoscale Science and Technology, award no. 70NANB14H209, through the University of Maryland. NIST disclaimer: selected commercial equipment, instruments or materials are identified to specify the adequacy of experimental conditions or reported results. Such identification does not imply recommendation or endorsement by the National Institute of Standards and Technology, nor does it imply that the equipment, instruments or materials identified are necessarily the best available for the purpose.

Author information

Authors and Affiliations

Authors

Contributions

C.R.L. and S.Q.T. conceived of and designed the study. C.R.L., N.L.M., G.L., E.C., Y.H., X.L., K.J., V.K., N.G.K., E.S. and C.T.P. performed experiments. Y.L., Y.Y., R.Z. and Y.H. performed computational analyses. S.M., G.K., J.M., Y.C. and S.Q.T. supervised the project. C.R.L. and S.Q.T. wrote the paper with input from all authors.

Corresponding author

Correspondence to Shengdar Q. Tsai.

Ethics declarations

Competing interests

C.R.L. and S.Q.T. have filed a patent application on CHANGE-seq. S.Q.T. is a co-inventor on patents covering CIRCLE-seq and GUIDE-seq. S.Q.T. is a member of the scientific advisory board of Kromatid.

Additional information

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

Extended data

Extended Data Fig. 1 Detailed overview of CHANGE-seq method.

Genomic DNA is randomly tagmented to an average of ~400 bp with a custom Tn5-transposome with an uracil-containing adapter. 9-nt Tn5-generated gaps in the DNA are filled in with a high-fidelity uracil-tolerant U+ polymerase and sealed with Taq DNA ligase. 4 bp overhangs are released with a mixture of USER enzyme and T4 PNK. DNA molecules are circularized at low concentrations that favor intramolecular ligation. Unwanted linear DNA is degraded with an exonuclease cocktail (comprised of Exonuclease I, Lambda exonuclease and Plasmid-Safe ATP-dependent DNase). Purified circular DNA is treated with Cas9:sgRNA RNP and cleaved DNA ends at on- and off-target sites are released for NGS library preparation, PCR amplification, and pair-end high-throughput sequencing.

Extended Data Fig. 2 Schematic comparison of CIRCLE-seq and CHANGE-seq workflows.

CHANGE-seq eliminates the requirement for specialized equipment for physical DNA shearing along with 9 additional enzymatic or purification steps. The simplified workflow substantially streamlines the process, decreases the requirement of input genomic DNA for circularization by approximately 5-fold and reduces the number of reactions to process each sample by 10- to 20-fold to a single reaction per sample.

Extended Data Fig. 3 CHANGE-seq detects all or nearly all sites detected by GUIDE-seq.

Venn diagrams showing the number of overlapping off-target sites captured by CHANGE-seq (blue) and GUIDE-seq (clear). The top six comparisons are of standard targets; the bottom four comparisons are of repetitive targets commonly used to benchmark genome-wide off-target activity detection methods.

Extended Data Fig. 4 GUIDE-seq optimization for human primary CD4+/CD8+ T cells.

a, Viability of cell population assessed by FACS analysis with DAPI staining 3 days post nucleofection with dsODN with phosphorothioate modifications at 5’ end, 3’ end, both ends or without dsODN (n=3). b, Indel rates at the intended target sites 3 days post nucleofection with dsODN with phosphorothioate modifications modifications at 5’ end, 3’ end, both ends or without dsODN (n=3). c, Integration rates of dsODNs with phosphorothioate modifications at 5’ end, 3’ end, both ends or without dsODN (n=3). d, Viability of cell population assessed by FACS analysis with DAPI staining 3 days post nucleofection with different doses of dsODN with 3’ end modifications (n=3). e, Indels rates at the intended target sites 3 days post nucleofection with different doses of dsODN with 3’ end modifications (n=3). f, dsODN integration rates 3 days post nucleofection with different doses of dsODN with 3’ end modifications (n=3). g, Scatterplots of GUIDE-seq read counts (log scale) between two independently prepared GUIDE-seq libraries for 3 target sites, showing GUIDE-seq technical reproducibility. Correlation between two samples was calculated using Pearson’s correlation coefficient.

Extended Data Fig. 5 Detailed characterization of a specific and active sgRNA targeting the TRAC region.

a, Manhattan plot showing the genome-wide distribution of sites identified in vitro by CHANGE-seq (arrow indicates the on-target site). b, Visualization of sites detected by CHANGE-seq. The intended target sequence is shown in the top line. Cleaved sites (on- and off-target) are shown underneath and are ordered top to bottom by CHANGE-seq read count, with mismatches to the intended target sequence indicated by colored nucleotides. Note that output is truncated to top sites with a full listing in Supplementary Table 3. c, Manhattan plot showing the on-target site detected for TRAC site 3 by GUIDE-seq, with no off-target sites being identified (arrow indicates the on-target site). d, Visualization of sites detected by GUIDE-seq. e, Indels rates at the intended target site 3 days post nucleofection (n=3). f, Flow plot showing distribution of TCR αβ expression in control (red) versus cells edited with sgRNA targeting TRAC site 3 (light blue). These experiments were performed three times with similar results. g, Barplot showing the percentage of TCR disruption 14 days after nucleofection with sgRNA:Cas9 complex measured by flow cytometry analysis (n=3).

Extended Data Fig. 6 GUIDE-seq dsODN tag independent indel frequencies are strongly correlated with tag integration frequencies.

Comparison of standard targeted sequencing and rhAMPSeq, a multiplex targeted sequencing method used in our study to validate on- and off-target site mutations. Scatterplots of indel mutation frequencies (top) and tag integration frequencies (bottom), between standard amplicon sequencing and rhAMPSeq, for sgRNAs targeted against CTLA4 site 9 and TRAC site 2 (See Methods). b, Scatterplots showing correlation between indel frequencies (in cells edited with Cas9 RNPs and no dsODN tag) and tag integration frequencies (in cells edited with Cas9 RNP and dsODN tag) at on- and off-target sites measured by targeted amplicon sequencing. (a-b) Correlation between two samples was calculated using Pearson’s correlation coefficient.

Extended Data Fig. 7 Targeted tag sequencing validation of CHANGE-seq detected off-target sites.

Targeted tag integration frequencies evaluated by standard targeted sequencing (triangle shape) and or rhAMPSeq (circle shape) (See Methods) at on- and off-target sites detected by both GUIDE-seq and CHANGE-seq, or detected by CHANGE-seq only (classes A-D), for sgRNAs targeted to TRAC site 2 and CTLA4 site 9. Panels for sites identified by both GUIDE-seq and CHANGE-seq and classes A and B for TRAC site 2 duplicated from main Fig. 4f for completeness.

Extended Data Fig. 8 GUIDE-seq read counts are strongly correlated with indel and tag integration frequencies in human primary T cells.

a, Scatterplots showing correlation between indel frequencies and GUIDE-seq read counts at on- and off-target sites, and b, tag integration and GUIDE-seq read counts at on- and off-target sites. (a-b) Correlation between two samples was calculated using Pearson’s correlation coefficient.

Extended Data Fig. 9 Influence of chromatin state on CRISPR-Cas9 genome-wide off-target activity.

a, Barplots showing the enrichment of individual epigenetic feature in GUIDE-seq (n=1,196), CHANGE-seq (n=11,000) and Cas-OFFinder (n=11,000). Statistical significance was calculated using two-tailed Welch’s t-test. Error bars indicate 95% confidence interval, estimated from 1000 bootstrap samples. b, Heatmap showing fold enrichment for various genomic annotations computed by ChromHMM for validation of chromatin state annotations. Darker colors represent higher fold enrichment.

Extended Data Fig. 10 CHANGE-seq enables detection of effects of individual genetic variation on genome-wide activity of genome editors.

a, Scatterplots of CHANGE-seq read counts (log scale) between two CHANGE-seq libraries independently prepared from the same source of genomic DNA, evaluating 6 target sites in 7 different genomes, showing that CHANGE-seq is highly reproducible. Correlation between two samples was calculated using Pearson’s correlation coefficient. b, Pairwise M/A plots for visualizing read count differences. The ratio (M) versus the average (A) of CHANGE-seq read counts (log scale) performed on the indicated GIAB or human T-cell sample versus a GM12878 GIAB reference sample. Each point represents an off-target site, and off-target sites that contain a non-reference single-nucleotide variant (SNV) are labelled in red.

Supplementary information

Supplementary Information

Supplementary Protocol, Supplementary Note, Supplementary Table 1, Supplementary Figures 1–3.

Reporting Summary

Supplementary Tables 2–8

This file contains Supplementary Tables 2–8. Supplementary Table 2 contains sgRNA target sites and NGS primers list; Supplementary Table 3 contains complete list of CHANGE-seq-detected sites; Supplementary Table 4 contains a list of CHANGE-seq target site specificity ratios; Supplementary Table 5 contains a list of on-target site targeted sequencing counts; Supplementary Table 6 contains a complete list of GUIDE-seq-detected sites; Supplementary Table 7 contains a list of off-target site targeted sequencing counts (standard targeted sequencing); Supplementary Table 8 contains a list of off-target site targeted sequencing counts (rhAmpSeq).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lazzarotto, C.R., Malinin, N.L., Li, Y. et al. CHANGE-seq reveals genetic and epigenetic effects on CRISPR–Cas9 genome-wide activity. Nat Biotechnol 38, 1317–1327 (2020). https://doi.org/10.1038/s41587-020-0555-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-020-0555-7

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