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.

  • Brief Communication
  • Published:

A rationally engineered cytosine base editor retains high on-target activity while reducing both DNA and RNA off-target effects

Abstract

Cytosine base editors (CBEs) offer a powerful tool for correcting point mutations, yet their DNA and RNA off-target activities have caused concerns in biomedical applications. We describe screens of 23 rationally engineered CBE variants, which reveal mutation residues in the predicted DNA-binding site can dramatically decrease the Cas9-independent off-target effects. Furthermore, we obtained a CBE variant—YE1-BE3-FNLS—that retains high on-target editing efficiency while causing extremely low off-target edits and bystander edits.

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: On-target efficiency of engineered CBEs.
Fig. 2: DNA and RNA off-target evaluation of engineered CBEs.
Fig. 3: Activities of engineered BE3-FNLS and BE3-hA3A.

Similar content being viewed by others

Data availability

All the sequencing data were deposited in NCBI Sequence Read Archive (SRA) under project accession PRJNA527003 and https://www.biosino.org/node/project/detail/OEP000272. The data that support the findings of this study are available from the corresponding author upon request.

References

  1. 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).

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

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

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

    Article  CAS  Google Scholar 

  5. Grünewald, J. et al. CRISPR adenine and cytosine base editors with reduced RNA off-target activities. 37, 1041–1048 (2019).

  6. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 37, 1041–1048 (2019).

  7. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371–376 (2017).

    Article  CAS  Google Scholar 

  8. Holden, L. G. et al. Crystal structure of the anti-viral APOBEC3G catalytic domain and functional implications. Nature 456, 121–124 (2008).

    Article  CAS  Google Scholar 

  9. Chen, K. M. et al. Structure of the DNA deaminase domain of the HIV-1 restriction factor APOBEC3G. Nature 452, 116–119 (2008).

    Article  CAS  Google Scholar 

  10. Teng, B. B. et al. Mutational analysis of apolipoprotein B mRNA editing enzyme (APOBEC1). structure-function relationships of RNA editing and dimerization. J. Lipid Res. 40, 623–635 (1999).

    CAS  PubMed  Google Scholar 

  11. Teng, B., Burant, C. F. & Davidson, N. O. Molecular cloning of an apolipoprotein B messenger RNA editing protein. Science 260, 1816–1819 (1993).

    Article  CAS  Google Scholar 

  12. Gehrke, J. M. et al. An APOBEC3A–Cas9 base editor with minimized bystander and off-target activities. Nat Biotechnol. 36, 977–982 (2018).

  13. Wang, X. et al. Efficient base editing in methylated regions with a human APOBEC3A-Cas9 fusion. Nat Biotechnol. 36, 946–949 (2018).

    Article  CAS  Google Scholar 

  14. Zafra, M. P. et al. Optimized base editors enable efficient editing in cells, organoids and mice. Nat Biotechnol. 36, 888–893 (2018).

  15. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36, 843–846 (2018).

    Article  CAS  Google Scholar 

  16. 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  Google Scholar 

  17. Komor, A. C. et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci. Adv. 3, eaao4774 (2017).

  18. Doman, J. L., Raguram, A., Newby, G. A. & Liu, D. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat. Biotechnol. (in the press).

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

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  23. 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  Google Scholar 

  24. Wang, X. et al. CRISPR–DAV: CRISPR NGS data analysis and visualization pipeline. Bioinformatics 33, 3811–3812 (2017).

    Article  CAS  Google Scholar 

  25. Cibulskis, K. et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 31, 213–219 (2013).

    Article  CAS  Google Scholar 

  26. Wilm, A. et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets. Nucleic Acids Res. 40, 11189–11201 (2012).

    Article  CAS  Google Scholar 

  27. Saunders, C. T. et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 28, 1811–1817 (2012).

  28. Fang, H. et al. Indel variant analysis of short-read sequencing data with Scalpel. Nat. Protoc. 11, 2529–2548 (2016).

    Article  CAS  Google Scholar 

  29. 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  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).

    Article  Google Scholar 

  31. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

  34. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  35. Chen, C. C., Hwang, J. K. & Yang, J. M. (PS)2: protein structure prediction server. Nucleic Acids Res. 34, W152–157 (2006).

  36. Huang, T. T. et al. (PS)2: protein structure prediction server version 3.0. Nucleic Acids Res. 43, W338–342 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the FACS facility staff H. Wu, L. Quan and S. Qian at ION, M. Zhang at IPS, and L. Yuan at Big Data Platform (SIBS, CAS). This study was supported by the R&D Program of China (2018YFC2000100 and 2017YFC1001302, 2017YFC0909701), the CAS Strategic Priority Research Program (XDB32060000, XDBS01060100), the National Natural Science Foundation of China (31871502, 31522037, 31822035, 31922048, 31925016, 91957122), the Basic Frontier Scientific Research Program of Chinese Academy of Sciences From 0 to 1 original innovation project (ZDBS-LY-SM001), the Shanghai Municipal Science and Technology Major Project (2018SHZDZX05), the Shanghai City Committee of science and technology project (18411953700, 18JC1410100, 16JC1420202), the National Science and Technology Major Project (2015ZX10004801-005), the National Key Research and Development Program of China (2017YFA0505500, 2016YFC0901704) the Agricultural Science and Technology Innovation Program to E.Z. and the International Partnership Program of Chinese Academy of Sciences (153D31KYSB20170059).

Author information

Authors and Affiliations

Authors

Contributions

E.Z. designed and performed experiments. Y.S., W.W., R.Z. and Y.L. performed data analysis. T.Y., B.H. and J.L. performed PCR analysis. W.Y. performed mouse embryo transfer. C.Z. performed cell transfection experiments. H.Y. and Y.L. supervised the project and designed experiments. Y.S. and H.Y. wrote the paper.

Corresponding authors

Correspondence to Erwei Zuo, Yixue Li or Hui Yang.

Ethics declarations

Competing interests

The authors disclose a patent application relating to aspects of this work (engineered base editors).

Additional information

Peer review information Lei, Tang was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 The on-target editing ofthe BE3 and BE3 variants in different target sites.

a, The mutated residues are highlighted in the predicted structure of rAPOBEC1. Green and yellow colors indicate residues in the helix and the loop of the structure, respectively. b, The crystal structure of APOBEC3G. c, The on-target efficiency and indel frequencies of different versions of engineered CBEs for additional 11 target sites. d, The C-to-T editing efficiency for the engineered variants at each C of the 21 target sites. n = 21 independent experiments for each group. All values are presented as mean ± s.e.m. e, The indel frequency comparison among the engineered variants for the 21 target sites. n = 21 independent experiments for each group. P value was calculated by two-sided Student’s t-tests. Box-and-whisker plots: center line indicates median, the bottom and top lines of the box represents the first quartile and third quartile of the values, respectively. The bottom and top of the vertical line represent the minimum and maximum value. f, The on-target C-to-T editing efficiency of engineered BE3 variants at each target site. n = 3 biologically independent samples for each group. P value was calculated by two-sided Student’s t-tests. Sequences of the on-target protospacers and primers were shown in Extended Data Table 5. The data for BE3 and YE1-BE3 are also used in Figs. 3a and 3d-g.

Extended Data Fig. 2 The embryonicdevelopment rates for BE3 and BE3 variants.

a, The blastocyst rate of BE3 and BE3 variants with sgRNA-D. All values are presented as mean ± s.e.m. b, The blastocyst rate for BE3-hA3A and BE3-FNLSwith additional sgRNAs. All values are presented as mean ± s.e.m. n = 3 biologically independent samples for each group.

Extended Data Fig. 3 On-target editing efficiency and characteristics of off-target SNVs of engineered CBEs.

a, On-target editing efficiency of BE3 and CBE variants from WGS data. Two BE3 embryos without sgRNAs were not shown as they have no target site. b, Comparison of C-to-T and G-to-A conversions between CBE variants-treated and Cre or BE3 groups. n = 3 biologically independent samples for Cre, n = 6 biologically independent samples for BE3, n = 12 biologically independent samples for BE3R126E, n = 3 biologically independent samples for BE3R132E, n = 8 biologically independent samples for YE1-BE3, and n = 3 biologically independent samples for FE1-BE3. Two Cre samples and six BE3 samples were derived from Zuo et al.22 and one Cre sample was newly generated in this study. All values are presented as mean ± s.e.m. P value was calculated by two-sided Student’s t-test.

Extended Data Fig. 4 Venn diagrams of SNVs detected in each embryo by WGS data using the indicated software tools.

a, SNVs identified in BE3R126E-treated embryos. b, SNVs identified in BE3R132E-treated embryos. c, SNVs identified in YE1-BE3-treated embryos. d, SNVs identified in FE1-BE3-treated embryos. e, SNVs identified in the newly generated Cre-treated embryo.

Extended Data Fig. 5 Characteristics of off-target SNVs of engineered CBEs.

a, The overlap among SNVs detected from our analysis with predicted off-targets sites by Cas-OFFinder and CRISPOR.

Extended Data Fig. 6 Editing rate of RNA off-targets for BE3 variants at 36 h post-transfection.

Editing rate of each variant across the chromosomes for each sample.

Extended Data Fig. 7 RNA off-target evaluation of engineered CBEs at 72h post-transfection.

a, The comparison of the total number of detected RNA off-target SNVs at 72 h post-transfection. n = 6 biologically independent samples for GFP, n = 9 biologically independent samples for BE3, n = 7 biologically independent samples for BE3R126E and n = 2 biologically independent samples for YE1-BE3 groups. All values are presented as mean ± s.e.m. P values above each bar were calculated by comparing with GFP group with two-sided Student’s t-tests. b, The distribution of mutation types for GFP, BE3, and BE3 variants-treated groups. c, Editing rate of RNA off-targets for BE3 variants at 72 h post-transfection.

Extended Data Fig. 8 On-target editing efficiency and off-target effects of BE3-FNLS and BE3-hA3A.

a, The C-to-T editing efficiency for the engineered variants at each C of the 21 target sites. n = 21 independent experiments for each group. All values are presented as mean ± s.e.m. The data for BE3 are also used in Fig. 3d. b, SNVs identified in BE3-hA3AY130F and YE1-BE3-FNLS-treated embryos. c, The overlap among SNVs detected from our analysis with predicted off-targets sites by Cas-OFFinder and CRISPOR. d, The distribution of mutation types of DNA off-target SNVs for BE3-hA3AY130F and YE1-BE3-FNLS-treated embryos. e, The distribution of mutation types of RNA off-target SNVs for BE3-hA3AY130F and YE1-BE3-FNLS-treated embryos. f, The expression level of APOBEC1 in BE3 and BE3-FNLS variants. n = 3 biologically independent samples for each group. Box-and-whisker plots: center line indicates median, the bottom and top lines of the box represents the first quartile and third quartile of the values, respectively. The bottom and top of the vertical line represent the minimum and maximum value. g, Editing rate of RNA off-targets for BE3 and BE3-FNLS variants at 36 h post-transfection. n = 3 biological replicates for each group. P value was calculated by two-sided Student’s t-test.

Extended Data Fig. 9 The comparation of BE3-FNLS, YE1-BE3-FNLS and BE4max.

a, The C-to-T editing efficiency for BE3-FNLS, YE1-BE3-FNLS and BE4max at indicated target sites. b, Indel frequency for BE3-FNLS, YE1-BE3-FNLS and BE4max at indicated target sites. Data are shown as mean values ± SEM for n = 3 biological replicates performed at the same time. P value was calculated by two-sided Student’s t-test.

Extended Data Fig. 10 Activities of CBE and CBE variants at the indicated Cas9-dependent off-target sites.

a, The Cas9-dependent off-target effects of the CBE and CBE variants. b, The comparison of editing frequencies of CBE and CBE variants at 34 potential off-target sites. P values were calculated by two sided Student’s t-tests, compared with YE1-BE3-FNLS group. Each cell represents the percentage of total sequencing reads with C to T conversion. n = 21 independent experiments for each group. Box-and-whisker plots: center line indicates median, the bottom and top lines of the box represents the first quartile and third quartile of the values, respectively. The bottom and top of the vertical line represent the minimum and maximum value. HEK293T cells were transfected with plasmids expressing BE3, BE3R126E, BE3R132E, YE1-BE3, FE1-BE3, BE3-hA3A, BE3-hA3AY130F, BE3-FNLS and YE1-BE3-FNLS and sgRNAs matching the indicated on-target sequence using Lipofectamine 3000. Three days after transfection, genomic DNA was extracted, amplified by PCR, and analyzed by high-throughput DNA sequencing at the on-target loci, plus the top ten known Cas9 off-target loci for these sgRNAs, as previously determined using the GUIDE-seq method23, 24 and ChIP-seq method25. Sequences of the on-target and off-target protospacers and primers were shown in Extended Data Table 5.

Supplementary information

Supplementary Information

Supplementary Figure 1, Supplementary Tables 1–3, 5 and 6, and Supplementary Sequence 1

Reporting Summary

Supplementary Table

Allele sequence

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zuo, E., Sun, Y., Yuan, T. et al. A rationally engineered cytosine base editor retains high on-target activity while reducing both DNA and RNA off-target effects. Nat Methods 17, 600–604 (2020). https://doi.org/10.1038/s41592-020-0832-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-020-0832-x

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