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:

Split complementation of base editors to minimize off-target edits

Abstract

Base editors (BEs) empower the efficient installation of beneficial or corrective point mutations in crop and human genomes. However, conventional BEs can induce unpredictable guide RNA (gRNA)-independent off-target edits in the genome and transcriptome due to spurious activities of BE-enclosing deaminases, and current improvements mostly rely on deaminase-specific mutagenesis or exogenous regulators. Here we developed a split deaminase for safe editing (SAFE) system applicable to BEs containing distinct cytidine or adenosine deaminases, with no need of external regulators. In SAFE, a BE was properly split at a deaminase domain embedded inside a Cas9 nickase, simultaneously fragmenting and deactivating both the deaminase and the Cas9 nickase. The gRNA-conditioned BE reassembly conferred robust on-target editing in plant, human and yeast cells, while minimizing both gRNA-independent and gRNA-dependent off-target DNA/RNA edits. SAFE also substantially increased product purity by eliminating indels. Altogether, SAFE provides a generalizable solution for BEs to suppress off-target editing and improve on-target performance.

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: Development of the split CBE complementation system.
Fig. 2: Split-AID10 exhibits improved on-target performance.
Fig. 3: Split-AID10 exhibits minimal off-target effects.
Fig. 4: Expansion of the SAFE system to CBEs with APOBEC deaminases.
Fig. 5: Expansion of the SAFE system to a TadA8e-based ABE.
Fig. 6: Split-ABE8e exhibits minimal gRNA-independent off-target effects.

Similar content being viewed by others

Data availability

The amplicon data, WGS data and RNA-seq data reported in this paper can be found at the CNGB Sequence Archive of the China National GeneBank DataBase with the accession number CNP0004043. Amino acid sequences of different SAFE BEs and target amplicon sequences for the gRNAs are provided in the Supplementary Information. Source data are provided with this paper.

References

  1. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  3. 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 

  4. Kang, B. C. et al. Precision genome engineering through adenine base editing in plants. Nat. Plants 4, 427–431 (2018).

    CAS  PubMed  Google Scholar 

  5. Kim, J. S. Precision genome engineering through adenine and cytosine base editing. Nat. Plants 4, 148–151 (2018).

    CAS  PubMed  Google Scholar 

  6. Zhang, H. et al. Genome editing of upstream open reading frames enables translational control in plants. Nat. Biotechnol. 36, 894–898 (2018).

    CAS  PubMed  Google Scholar 

  7. Xue, C., Zhang, H., Lin, Q., Fan, R. & Gao, C. Manipulating mRNA splicing by base editing in plants. Sci. China Life Sci. 61, 1293–1300 (2018).

    CAS  PubMed  Google Scholar 

  8. Chen, K., Wang, Y., Zhang, R., Zhang, H. & Gao, C. CRISPR/Cas genome editing and precision plant breeding in agriculture. Annu. Rev. Plant Biol. 70, 667–697 (2019).

    CAS  PubMed  Google Scholar 

  9. Li, Z., Xiong, X., Wang, F., Liang, J. & Li, J. F. Gene disruption through base editing-induced messenger RNA missplicing in plants. New Phytol. 222, 1139–1148 (2019).

    CAS  PubMed  Google Scholar 

  10. Molla, K. A., Sretenovic, S., Bansal, K. C. & Qi, Y. Precise plant genome editing using base editors and prime editors. Nat. Plants 7, 1166–1187 (2021).

    CAS  PubMed  Google Scholar 

  11. Xue, C. et al. Tuning plant phenotypes by precise, graded downregulation of gene expression. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01707-w (2023).

  12. Rees, H. A. et al. Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat. Commun. 8, 15790 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. 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).

    CAS  PubMed  Google Scholar 

  14. Liang, Z. et al. Genome editing of bread wheat using biolistic delivery of CRISPR/Cas9 in vitro transcripts or ribonucleoproteins. Nat. Protoc. 13, 413–430 (2018).

    CAS  PubMed  Google Scholar 

  15. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Grünewald, J. et al. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat. Biotechnol. 37, 1041–1048 (2019).

    PubMed  PubMed Central  Google Scholar 

  22. 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).

    PubMed  PubMed Central  Google Scholar 

  23. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Jin, S. et al. Rationally designed APOBEC3B cytosine base editors with improved specificity. Mol. Cell 79, 728–740 (2020).

    CAS  PubMed  Google Scholar 

  25. Richter, M. F. et al. Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat. Biotechnol. 38, 883–891 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Neugebauer, M. E. et al. Evolution of an adenine base editor into a small, efficient cytosine base editor with low off-target activity. Nat. Biotechnol. 41, 673–685 (2023).

    CAS  PubMed  Google Scholar 

  28. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. Nat. Biotechnol. 41, 663–672 (2023).

    CAS  PubMed  Google Scholar 

  29. Wang, L. et al. Eliminating base-editor-induced genome-wide and transcriptome-wide off-target mutations. Nat. Cell Biol. 23, 552–563 (2021).

    PubMed  Google Scholar 

  30. Berríos, K. N. et al. Controllable genome editing with split-engineered base editors. Nat. Chem. Biol. 17, 1262–1270 (2021).

    PubMed  PubMed Central  Google Scholar 

  31. Liu, Y. et al. A Cas-embedding strategy for minimizing off-target effects of DNA base editors. Nat. Commun. 11, 6073 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Li, S. et al. Docking sites inside Cas9 for adenine base editing diversification and RNA off-target elimination. Nat. Commun. 11, 5827 (2020).

    PubMed  PubMed Central  Google Scholar 

  33. Nguyen Tran, M. T. et al. Engineering domain-inlaid SaCas9 adenine base editors with reduced RNA off-targets and increased on-target DNA editing. Nat. Commun. 11, 4871 (2020).

    PubMed  PubMed Central  Google Scholar 

  34. Liu, K. et al. Mapping single-cell-resolution cell phylogeny reveals cell population dynamics during organ development. Nat. Methods 18, 1506–1514 (2021).

    CAS  PubMed  Google Scholar 

  35. Wang, Y., Zhou, L., Liu, N. & Yao, S. BE-PIGS: a base-editing tool with deaminases inlaid into Cas9 PI domain significantly expanded the editing scope. Signal Transduct. Target. Ther. 4, 36 (2019).

    PubMed  PubMed Central  Google Scholar 

  36. Xiong, X. et al. A cytosine base editor toolkit with varying activity windows and target scopes for versatile gene manipulation in plants. Nucleic Acids Res. 50, 3565–3580 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Pham, P. et al. Structural analysis of the activation-induced deoxycytidine deaminase required in immunoglobulin diversification. DNA Repair 43, 48–56 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zong, Y. et al. Precise base editing in rice, wheat and maize with a Cas9–cytidine deaminase fusion. Nat. Biotechnol. 35, 438–440 (2017).

    CAS  PubMed  Google Scholar 

  39. Wang, Z. P. et al. Egg cell-specific promoter-controlled CRISPR/Cas9 efficiently generates homozygous mutants for multiple target genes in Arabidopsis in a single generation. Genome Biol. 16, 144 (2015).

    PubMed  PubMed Central  Google Scholar 

  40. Chen, Y. et al. CRISPR/Cas9-mediated base-editing system efficiently generates gain-of-function mutations in Arabidopsis. Sci. China Life Sci. 60, 520–523 (2017).

    CAS  PubMed  Google Scholar 

  41. Powles, S. B. & Yu, Q. Evolution in action: plants resistant to herbicides. Annu. Rev. Plant Biol. 61, 317–347 (2010).

    CAS  PubMed  Google Scholar 

  42. Jin, S., Gao, Q. & Gao, C. An unbiased method for evaluating the genome-wide specificity of base editors in rice. Nat. Protoc. 16, 431–457 (2021).

    CAS  PubMed  Google Scholar 

  43. DiCarlo, J. E. et al. Genome engineering in Saccharomyces cerevisiae using CRISPR–Cas systems. Nucleic Acids Res. 41, 4336–4343 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Long, H. et al. Antibiotic treatment enhances the genome-wide mutation rate of target cells. Proc. Natl Acad. Sci. USA 113, E2498–E2505 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Ren, Q. et al. Improved plant cytosine base editors with high editing activity, purity, and specificity. Plant Biotechnol. J. 19, 2052–2068 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Randall, L. B. et al. Genome- and transcriptome-wide off-target analyses of an improved cytosine base editor. Plant Physiol. 187, 73–87 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Li, S., Liu, L., Sun, W., Zhou, X. & Zhou, H. A large-scale genome and transcriptome sequencing analysis reveals the mutation landscapes induced by high-activity adenine base editors in plants. Genome Biol. 23, 51 (2022).

    PubMed  PubMed Central  Google Scholar 

  48. Zong, Y. et al. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A. Nat. Biotechnol. 36, 950–953 (2018).

    CAS  Google Scholar 

  49. Lapinaite, A. et al. DNA capture by a CRISPR–Cas9-guided adenine base editor. Science 369, 566–571 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Chen, L. et al. Engineering a precise adenine base editor with minimal bystander editing. Nat. Chem. Biol. 19, 101–110 (2023).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Jiang, F., Zhou, K., Ma, L., Gressel, S. & Doudna, J. A. A Cas9–guide RNA complex preorganized for target DNA recognition. Science 348, 1477–1481 (2015).

    CAS  PubMed  Google Scholar 

  53. Jiang, F. et al. Structures of a CRISPR–Cas9 R-loop complex primed for DNA cleavage. Science 351, 867–871 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kummerfeld, S. K. & Teichmann, S. A. Relative rates of gene fusion and fission in multi-domain proteins. Trends Genet. 21, 25–30 (2005).

    CAS  PubMed  Google Scholar 

  55. Chee, W. K. D., Yeoh, J. W., Dao, V. L. & Poh, C. L. Highly reversible tunable thermal-repressible split-T7 RNA polymerases (Thermal-T7RNAPs) for dynamic gene regulation. ACS Synth. Biol. 11, 921–937 (2022).

    CAS  PubMed  Google Scholar 

  56. 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).

    PubMed  PubMed Central  Google Scholar 

  57. Wang, L. et al. Enhanced base editing by co-expression of free uracil DNA glycosylase inhibitor. Cell Res. 27, 1289–1292 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Kim, H. S., Jeong, Y. K., Hur, J. K., Kim, J. S. & Bae, S. Adenine base editors catalyze cytosine conversions in human cells. Nat. Biotechnol. 37, 1145–1148 (2019).

    CAS  PubMed  Google Scholar 

  59. Wu, Y. et al. Genome-wide analyses of PAM-relaxed Cas9 genome editors reveal substantial off-target effects by ABE8e in rice. Plant Biotechnol. J. 20, 1670–1682 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Li, Z. et al. A potent Cas9-derived gene activator for plant and mammalian cells. Nat. Plants 3, 930–936 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Clough, S. J. & Bent, A. F. Floral dip: a simplified method for Agrobacterium-mediated transformation of Arabidopsis thaliana. Plant J. 16, 735–743 (1998).

    CAS  PubMed  Google Scholar 

  62. Nishimura, A., Aichi, I. & Matsuoka, M. A protocol for Agrobacterium-mediated transformation in rice. Nat. Protoc. 1, 2796–2802 (2006).

    CAS  PubMed  Google Scholar 

  63. Liu, Q. et al. Hi-TOM: a platform for high-throughput tracking of mutations induced by CRISPR/Cas systems. Sci. China Life Sci. 62, 1–7 (2019).

    PubMed  Google Scholar 

  64. Gietz, R. D. & Schiestl, R. H. Quick and easy yeast transformation using the LiAc/SS carrier DNA/PEG method. Nat. Protoc. 2, 35–37 (2007).

    CAS  PubMed  Google Scholar 

  65. Lõoke, M., Kristjuhan, K. & Kristjuhan, A. Extraction of genomic DNA from yeasts for PCR-based applications. BioTechniques 50, 325–328 (2011).

    PubMed  PubMed Central  Google Scholar 

  66. Hwang, G. H. et al. Web-based design and analysis tools for CRISPR base editing. BMC Bioinform. 19, 542 (2018).

    CAS  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Xie, X. et al. CRISPR-GE: a convenient software toolkit for CRISPR-based genome editing. Mol. Plant 10, 1246–1249 (2017).

    CAS  PubMed  Google Scholar 

  69. 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 

  70. 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 

  71. Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Shen, M. W. et al. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature 563, 646–651 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key Research and Development Program of China grant no. 2019YFA0906202, the Guangdong Provincial Key Project for Basic and Applied Basic Research (Cornerstone grant) and the National Natural Science Foundation of China (NSFC) grant no. 32125004 to J.-F.L.; NSFC grant no. 32293191 to X.H.; NSFC grant no. 32200494 and the China Postdoctoral Science Foundation grant no. 2022M723662 to K.L.; and NSFC grant no. 32100485 to F.-N.X. We thank Azenta Life Sciences (Suzhou, China) for help in deep sequencing and Y. Chen for assistance in flow cytometry.

Author information

Authors and Affiliations

Authors

Contributions

J.-F.L. and K.L. conceived and designed the study. X.X., K.L., Z.L., F.-N.X. and X.-M.R. performed the experiments. K.L. and X.X. analysed the data. J.-F.L. and X.H. supervised the research. J.-F.L. wrote the manuscript. All authors approved the final version of the paper.

Corresponding authors

Correspondence to Kehui Liu or Jian-Feng Li.

Ethics declarations

Competing interests

X.X., K.L., Z.L., X.H. and J.-F.L. have been granted a China invention patent (ZL202210503831.5) based on some results reported in this paper. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Plants thanks Seiichi Toki and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 N- or C-terminal split fragment of PIGS-AID10 alone is catalytically inactive.

a, PIGS-AID10 exhibits higher editing efficiency than N-AID10. Comparison of C-to-T editing efficiencies between PIGS-AID10 and N-AID10 was conducted at indicated Arabidopsis endogenous genomic loci in protoplasts by deep sequencing of target amplicons. Data are shown as mean values of two biological replicates. Ctrl corresponds to cells transfected with the gRNA only. b, Amino acid sequence of AID10 with seven candidate split sites highlighted in red and the catalytic center shadowed in blue. c,d, None of the N-terminal (c) or C-terminal (d) split fragments of PIGS-AID10 alone can induce CBE reporter activity in the presence of gRNA. The gRNA only was used as a negative control (Ctrl). Relative luciferase (LUC) activity was calculated by setting the LUC activity of the unsplit CBE (PIGS-AID10) as 100%. Data are shown as mean values of two biological replicates.

Extended Data Fig. 2 Split-AID10 can edit the AtALS locus to confer herbicide resistance.

a, The AtALS target locus. Intended C·G to T·A (red) conversion by Split-AID10 would introduce an A-to-T amino acid substitution. The PAM is in green. b, Summary of Split-AID10 editing outcome in transgenic Arabidopsis T1 plants. The editing efficiency was calculated as the percentage of transgenic plants containing indicated mutation type at the target site. WT, Ho, He, Bi, and Chi denote wild type, homozygous, heterozygous, biallelic, and chimeric plants, respectively. c, Sanger sequencing validates homozygous C-to-T mutation introduced by Split-AID10. d, Homozygous C-to-T mutation at the AtALS locus confers the plant with imazethapyr herbicide resistance. Transgenic Arabidopsis T1 plants of 40-day-old were sprayed with 30 mg/l imazethapyr (IMZ) herbicide once and were photographed in one month. Unedited plants died of imazethapyr toxicity.

Extended Data Fig. 3 Split-AID10 enables robust on-target editing in human and yeast cells.

a,c, Split-AID10 exhibits sufficiently high on-target editing efficiency in human HEK293T cells (a) and yeast cells (c). Data are shown as mean values and standard deviation of three biological replicates for six target loci and the highest C-to-T editing efficiency among multiple Cs was used to represent the editing efficiency at a given target locus. b,d, Split-AID10 minimizes C-to-A/G and indel byproducts in human HEK293T cells (b) and yeast cells (d). The inlaid panel in d corresponds to a magnified view. Each dot in the violin plot represents the editing efficiency for the indicated editing product per target site, while the three lines mark quartile positions. e, Domain structures of Split-AID10N6, Split-AID10C6, and PIGS-AID10N6. Note that PIGS-AID10N6 contains a complete nCas9 and an intact deaminase catalytic center. f, None of Split-AID10N6, Split-AID10C6 and PIGS-AID10N6 is catalytically active in yeast cells. Data are shown as mean values and standard deviation of three biological replicates.

Extended Data Fig. 4 Rationale of visible selection of on-target editing in the EASY assay.

The intended nonsense mutation in ScAde1 or ScCan1 by converting a Trp or Gln codon (shadowed in yellow) to a stop codon via C-to-T editing (red) makes edited yeast clones visually distinguishable. The on-target C-to-T editing of ScAde1 generated red colonies in YPD agar plates, while that of ScCan1 produced white colonies in SC-Arg agar plates containing L-Canavanine.

Extended Data Fig. 5 Split-AID10 minimizes off-target DNA edits in human cells.

a,b, R-loop assay reveals that Split-AID10 eliminates unguided DNA edits relative to other tested CBEs in HEK293T cells. In a, data are shown as mean values and standard deviation of three biological replicates. In b, each dot in the violin plot represents the C-to-T editing efficiency per target site, while the three lines mark quartile positions. ns, not significant, two-sided Mann-Whitney U test. c, Split-AID10 induces substantially reduced edits at predicted off-target sites for the HEK-1 or HEK-5 targeting gRNA in HEK293T cells. Data are shown as mean values and standard deviation of three biological replicates.

Extended Data Fig. 6 Split-AID10 eliminates off-target RNA edits in human cells.

a,b, Gating strategies of flow cytometry for sorting base editor-expressing HEK293T cells and control cells. In a, cells expressing base editors were flow-sorted for the top 5% of gated cells (% parent) with the highest GFP (488 nm) and mScarlet (561 nm) signal. In b, the nCas9-UGI control was sorted for the top 5% of gated cells with the highest signal and for a population of cells with a mean fluorescence intensity matching the top 5% of BE3-transfected cells collected on the same day. c, On-target DNA editing validation for all base editor groups in HEK293T cell-based RNA off-target assay. Data are shown for three biological replicates (Rep.1-Rep.3). The lowercase number indicates the position of individual Cs in the protospacer (counting PAM as 21-23). d, Representative Manhattan plot (Rep.3) for transcriptome-wide off-target C-to-U edits in individual chromosomes in each group. n, total number of C-to-U edits. Dashed lines indicate a C-to-U editing level of 5%.

Extended Data Fig. 7 Split-BE3 eliminates off-target RNA edits in human cells.

Representative Manhattan plot (Rep.2) for transcriptome-wide off-target C-to-U edits in individual chromosomes in each group. n, total number of C-to-U edits. Dashed lines indicate a C-to-U editing level of 5%.

Extended Data Fig. 8 Split-ABE8e minimizes off-target RNA/DNA edits.

a,b, None of the N-terminal (a) or C-terminal (b) split fragments of PIGS-ABE8e alone can induce ABE reporter activity in protoplasts in the presence of gRNA. The gRNA only was used as a negative control (Ctrl). Relative luciferase (LUC) activity was calculated by setting the LUC activity of the unsplit ABE (PIGS-ABE8e) as 100%. Data are shown as mean values of two biological replicates. c, Representative Manhattan plot (Rep.3) for transcriptome-wide off-target A-to-I edits in individual chromosomes in each group in human HEK293T cell-based RNA off-target assay. n, total number of A-to-I edits. Dashed lines indicate a A-to-I editing level of 5%. d, Split-ABE8e induces substantially reduced edits at predicted off-target sites for the HEK-8 targeting gRNA in HEK293T cells. Data are shown as mean values and standard deviation of three biological replicates.

Extended Data Fig. 9 Split fragments of a BE are expressed at higher levels than the unsplit BE.

a, Split-AID10N6 and Split-AID10C6 are expressed at higher levels than PIGS-AID10 in both rice and Arabidopsis protoplasts. b, Split-ABE8eN6 and Split-ABE8eC6 are expressed at higher levels than PIGS-ABE8e in both rice and Arabidopsis protoplasts. In (a) and (b), 200 µL rice or Arabidopsis protoplasts were transfected with equal amounts of plasmid(s) encoding PIGS-BE alone or Split-BE-N plus Split-BE-C. Since Split-BE-N and Split-BE-C carry the same protein tag (2×HA tag for both PIGS-AID10 split fragments and 2×FLAG tag for both PIGS-ABE8e split fragments), Split-BE-N or Split-BE-C was also expressed alone to help distinguish individual products. A plasmid encoding GFP was co-transfected as an internal transfection control. The experiments were conducted twice with similar results. The white, orange, and blue arrowheads mark PIGS-BE, Split-BE-N, and Split-BE-C, respectively. Rubisco staining indicates equal protein loading.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1–5 and Sequences.

Reporting Summary

Source data

Source Data Extended Data Fig. 9

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xiong, X., Liu, K., Li, Z. et al. Split complementation of base editors to minimize off-target edits. Nat. Plants 9, 1832–1847 (2023). https://doi.org/10.1038/s41477-023-01540-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41477-023-01540-8

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