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:

Mitochondrial base editor induces substantial nuclear off-target mutations

Abstract

DddA-derived cytosine base editors (DdCBEs)—which are fusions of split DddA halves and transcription activator-like effector (TALE) array proteins from bacteria—enable targeted C•G-to-T•A conversions in mitochondrial DNA1. However, their genome-wide specificity is poorly understood. Here we show that the mitochondrial base editor induces extensive off-target editing in the nuclear genome. Genome-wide, unbiased analysis of its editome reveals hundreds of off-target sites that are TALE array sequence (TAS)-dependent or TAS-independent. TAS-dependent off-target sites in the nuclear DNA are often specified by only one of the two TALE repeats, challenging the principle that DdCBEs are guided by paired TALE proteins positioned in close proximity. TAS-independent off-target sites on nuclear DNA are frequently shared among DdCBEs with distinct TALE arrays. Notably, they co-localize strongly with binding sites for the transcription factor CTCF and are enriched in topologically associating domain boundaries. We engineered DdCBE to alleviate such off-target effects. Collectively, our results have implications for the use of DdCBEs in basic research and therapeutic applications, and suggest the need to thoroughly define and evaluate the off-target effects of base-editing tools.

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: DdCBE induces abundant off-target edits in the nuclear genome.
Fig. 2: DdCBE induces one-sided, TAS-dependent nDNA off-target edits.
Fig. 3: Prevalent, non-random TAS-independent off-target sites.
Fig. 4: TAS-independent nDNA off-target sites are enriched at CTCF binding sites and TAD boundaries.
Fig. 5: DdCBE variants with improved specificity.

Similar content being viewed by others

Data availability

All data generated for this paper have been deposited at NCBI Gene Expression Omnibus (GEO) and are available under GEO accession number GSE173859 (Detect-seq data), GSE173689 (ATAC-seq data and in situ ChIP–seq data) and GSE176089 (targeted deep sequencing data). hg38 was used as the reference genome. The Hi-C, DNase-seq, Bisulfite-seq and ChIP–seq data were downloaded from the GEO or ENCODE database; accession numbers of these public data sets are available in Supplementary Table 5.

Code availability

Detect-seq tools, including several Python scripts, were deposited on GitHub (https://github.com/menghaowei/Detect-seq). Detect-seq tools can help to perform Detect-seq analysis, including but not limited to Detect-seq signal finding, enrichment testing, off-target sites identification, TALE sequence alignment and alignment results visualization.

References

  1. Mok, B. Y. et al. A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature 583, 631–637 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Russell, O. M., Gorman, G. S., Lightowlers, R. N. & Turnbull, D. M. Mitochondrial diseases: hope for the future. Cell 181, 168–188 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Vafai, S. B. & Mootha, V. K. Mitochondrial disorders as windows into an ancient organelle. Nature 491, 374–383 (2012).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Stewart, J. B. & Chinnery, P. F. The dynamics of mitochondrial DNA heteroplasmy: implications for human health and disease. Nat. Rev. Genet. 16, 530–542 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Stewart, J. B. & Chinnery, P. F. Extreme heterogeneity of human mitochondrial DNA from organelles to populations. Nat. Rev. Genet. 22, 106–118 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Montano, V., Gruosso, F., Simoncini, C., Siciliano, G. & Mancuso, M. Clinical features of mtDNA-related syndromes in adulthood. Arch. Biochem. Biophys. 697, 108689 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Bacman, S. R. et al. MitoTALEN reduces mutant mtDNA load and restores tRNA(Ala) levels in a mouse model of heteroplasmic mtDNA mutation. Nat. Med. 24, 1696–1700 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bacman, S. R., Williams, S. L., Pinto, M., Peralta, S. & Moraes, C. T. Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs. Nat. Med. 19, 1111–1113 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Gammage, P. A., Rorbach, J., Vincent, A. I., Rebar, E. J. & Minczuk, M. Mitochondrially targeted ZFNs for selective degradation of pathogenic mitochondrial genomes bearing large-scale deletions or point mutations. EMBO Mol. Med. 6, 458–466 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lei, Z. et al. Detect-seq reveals out-of-protospacer editing and target-strand editing by cytosine base editors. Nat. Methods 18, 643–651 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Zhu, C. et al. Single-cell 5-formylcytosine landscapes of mammalian early embryos and ESCs at single-base resolution. Cell Stem Cell 20, 720–731.e725 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Shu, X. et al. Genome-wide mapping reveals that deoxyuridine is enriched in the human centromeric DNA. Nat. Chem. Biol. 14, 680–687 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Xia, B. et al. Bisulfite-free, base-resolution analysis of 5-formylcytosine at the genome scale. Nat. Methods 12, 1047–1050 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  16. 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  PubMed  PubMed Central  Google Scholar 

  17. Li, X. et al. Base editing with a Cpf1-cytidine deaminase fusion. Nat. Biotechnol. 36, 324–327 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Wang, X. et al. Cas12a base editors induce efficient and specific editing with low DNA damage response. Cell Rep. 31, 107723 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Sardo, L. et al. Real-time visualization of chromatin modification in isolated nuclei. J. Cell Sci. 130, 2926–2940 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, Q. et al. CoBATCH for high-throughput single-cell epigenomic profiling. Mol. Cell 76, 206–216.e207 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. Boch, J. et al. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326, 1509–1512 (2009).

    Article  ADS  CAS  PubMed  Google Scholar 

  22. Moscou, M. J. & Bogdanove, A. J. A simple cipher governs DNA recognition by TAL effectors. Science 326, 1501 (2009).

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Lamb, B. M., Mercer, A. C. & Barbas, C. F. III. Directed evolution of the TALE N-terminal domain for recognition of all 5′ bases. Nucleic Acids Res. 41, 9779–9785 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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 

  27. Nakahashi, H. et al. A genome-wide map of CTCF multivalency redefines the CTCF code. Cell Rep. 3, 1678–1689 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Schmidt, D. et al. Waves of retrotransposon expansion remodel genome organization and CTCF binding in multiple mammalian lineages. Cell 148, 335–348 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Merkenschlager, M. & Nora, E. P. CTCF and cohesin in genome folding and transcriptional gene regulation. Annu. Rev. Genomics Hum. Genet. 17, 17–43 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Rowley, M. J. & Corces, V. G. Organizational principles of 3D genome architecture. Nat. Rev. Genet. 19, 789–800 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Shi, Z., Gao, H., Bai, X. C. & Yu, H. Cryo-EM structure of the human cohesin–NIPBL–DNA complex. Science 368, 1454–1459 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Davidson, I. F. et al. DNA loop extrusion by human cohesin. Science 366, 1338–1345 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  33. Kim, Y., Shi, Z., Zhang, H., Finkelstein, I. J. & Yu, H. Human cohesin compacts DNA by loop extrusion. Science 366, 1345–1349 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Murayama, Y. & Uhlmann, F. Biochemical reconstitution of topological DNA binding by the cohesin ring. Nature 505, 367–371 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  35. Petela, N. J. et al. Scc2 Is a potent activator of cohesin’s ATPase that promotes loading by binding Scc1 without Pds5. Mol. Cell 70, 1134–1148.e1137 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hashimoto, H. et al. Structural basis for the versatile and methylation-dependent binding of CTCF to DNA. Mol. Cell 66, 711–720.e713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Yu, M. & Ren, B. The three-dimensional organization of mammalian genomes. Annu. Rev. Cell Dev. Biol. 33, 265–289 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Schmitt, A. D., Hu, M. & Ren, B. Genome-wide mapping and analysis of chromosome architecture. Nat. Rev. Mol. Cell Biol. 17, 743–755 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144–149 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yang, J. et al. ULtiMATE system for rapid assembly of customized TAL effectors. PLoS ONE 8, e75649 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21–29 (2015).

    PubMed Central  Google Scholar 

  44. Neely, A. E. & Bao, X. Nuclei isolation staining (NIS) method for imaging chromatin-associated proteins in difficult cell types. Curr. Protoc. Cell Biol. 84, e94 (2019).

    PubMed  PubMed Central  Google Scholar 

  45. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal https://doi.org/10.14806/ej.17.1.200 (2011).

  46. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  50. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wolff, J. et al. Galaxy HiCExplorer 3: a web server for reproducible Hi-C, capture Hi-C and single-cell Hi-C data analysis, quality control and visualization. Nucleic Acids Res. 48, W177–W184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank W. Wei for providing related plasmids; H. Cheng for sharing the antibodies for mitochondrial markers; W. Xie and X. Lu for discussion about the ChIP assay; X. Zhang and Chuyun Shao for help with ATAC-seq experiments and data processing; National Center for Protein Sciences at Peking University for assistance with FACS, imaging, sequencing, Imaris, Fragment Analyzer and Agilent 4150 TapeStation System; C. Shan, L. Fu, S. Qin and Y. Guo for assistance with immunofluorescence experiments, FACS and image processing; and G. Li and X. Zhang for assistance with NGS experiments. Bioinformatics analysis was performed on the High-Performance Computing Platform of the School of Life Sciences and High-Performance Computing Platform of the Center for Life Science. This work was supported by the National Natural Science Foundation of China (nos. 21825701, 91953201, 92153303 and 22107006), National Key R&D Program (2019YFA0110900 and 2019YFA0802200) and China Postdoctoral Science Foundation (2020M680218, 2021M700238). L. Liu was supported in part by the Postdoctoral Fellowship of Peking-Tsinghua Center for Life Sciences.

Author information

Authors and Affiliations

Authors

Contributions

Z.L., H.M. and C.Y. conceived and led the project. Z.L., H.M., L.L. and C.Y. designed the experiments to investigate the DdCBE off-target effect, which were performed by Z.L. and L.L. H.M. analysed Detect-seq, in situ ChIP–seq and the downloaded public data. H.Z. analysed ATAC-seq and targeted deep sequencing data. Z.L. performed the co-immunoprecipitation and non-fixation immunofluorescence assays. L.L. conducted the cell fractionation assay and the sequential western blot and fixation-based immunofluorescence experiments. X. R. and Y. Y. assisted the experiments and data processing. Z.L. and C.Y. designed the in situ ChIP experiments with the advice of A.H. M.L. performed the in situ ChIP–seq experiments with cell samples prepared by Z.L. Z.L., H.M., L.L., H.Z. and C.Y. wrote the paper with the help of X. R. and H.W.

Corresponding author

Correspondence to Chengqi Yi.

Ethics declarations

Competing interests

Peking University has filed patent applications on Detect-seq and optimized DdCBE variants described in this study, listing Z.L., H.M., Z.C.L., L.L, H.Z. and C.Y. as inventors.

Peer review

Peer review information

Nature thanks Bryan Dickinson and Fyodor Urnov 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 figures and tables

Extended Data Fig. 1 Workflow of Detect-seq.

Endogenous 5fdC was protected by O-ethylhydroxylamine (EtONH2). Damage repair step eliminates endogenous DNA damages including abasic sites (AP), single strand breaks (SSB), etc. Deoxyuridine (dU) generated by DdCBE in vivo was labeled by the in vitro reconstituted base excision repair (BER) reaction: UDG specifically recognizes and cleaves dU, leaving abasic sites; Endo IV removes abasic sites, leaving 3’-OH remnant; Bst DNA polymerase initiates DNA strand replacement after the 3’-OH; ligase sews the final nicks. Through the so-called “nick translation” activity of Bst polymerase during this step, biotinylated dUTPs and 5fdCTPs were incorporated 3’ to dU. Malononitrile treatment marks the incorporated 5fdCs, generating a characteristic tandem C-to-T mutation pattern to trace DdCBE edits. Biotin pulldown followed by NaOH treatment enriches DdCBE edited DNA fragments and enhances Detect-seq signals.

Extended Data Fig. 2 Comparisons of Detect-seq signals for off-target edits under two different transfection conditions.

The two conditions are: 4x105 seeded HEK293T cells on 6-well plates were transfected with 4 μg of each monomer using 12 μl Lipofectamine 2000; or, 6.4x105 cells were transfected with 3.5 μg of each monomer using 21 μl Lipofectamine LTX. The Detect-seq signals are highly consistent between the two conditions for all three DdCBEs.

Extended Data Fig. 3 Editing ratios of nuclear DNA off-target sites identified for the three L1397N-DdCBEs.

a–c, Targeted deep sequencing results for selected nuclear off-target sites of ND4-L1397N (a), ND5.1-L1397N (b) and ND6-L1397N (c). For each off-target site, the editing ratio for the highest edited cytosine is plotted (blue), and the matched ratio in untreated ctrl sample is plotted in gray.

Extended Data Fig. 4 A real-time IF staining assay using unfixed HeLa nuclei to demonstrate the nuclear localization of DdCBE.

a, Fluorescence imaging of DAPI (navy blue), HA-tagged left half (Anti-HA, orange red) and Flag-tagged right half (Anti-Flag, green) in unfixed nuclei of HeLa cells untreated or transfected with Lipofectamine LTX. Possible mitochondrial contamination was tested by MitoTracker (magenta). The images were obtained at a representative Z-axis under the same exposure condition by High Speed Spinning Disk Confocal Microscope (ANDOR). Scale bars, 3 μm. Images are representative of 3 independent biological replicates. b, c, The projected 2D fluorescence image (b) and 3D snapshot (c) of a representative nuclei from cells transfected with Lipofectamine 3000. d, e, The projected 2D fluorescence image (d) and 3D snapshot (e) of a representative nuclei from cells transfected with Lipofectamine LTX. f, Statistic diagram for 3D mean fluorescence intensity per voxel of all scanned nuclei under different treatments. The data in bf for each nucleus was obtained from z-stack images collected at 0.4 μm intervals under the same exposure condition by DeltaVision OMX SR (GE). Similar color and scale bars in a were used. HeLa cells on 6-well plates were transfected with 2 μg of each monomer using 6 μl Lipofectamine3000; or, cells were transfected with 3.5 μg of each monomer using 21 μl Lipofectamine LTX. “ND6-WT”: wild type ND6-L1397N; “ND6-(TALE-)”: ND6-L1397N architectures that deleted the TALE arrays. In f, error bars reflect the mean +/− SD; and p-values are calculated by one-side Student’s t-test.

Extended Data Fig. 5 A small portion of DdCBE is localized in the nucleus of transfected HEK293T cells.

a, Western blotting results showing the distribution of ND6-L1397N (WT) in different subcellular fractions of 2×103 HEK293T cells untreated or transfected using Lipofectamine 2000 or LTX; and the distribution of three deletion variants of ND6-L1397N in different fractions of cells transfected with LTX. “DddA-free”, “UGI-free” and “TALE-free” mean the deletion of DddA, UGI and TALE arrays from the full-length ND6-L1397N respectively. The results show that ND6-L1397N is partially localized in the chromatin fraction no matter which transfection reagent was used. The signal of the TALE-free construct in the chromatin fraction is only present when the exposure time is extended. This observation suggests that compared to DddA and UGI, the TALE arrays most strongly affect the nuclear localization. ATP5a (mitochondria), GAPDH (cytosolic) and H3 (chromatin) were chosen as compartment-specific markers, demonstrating the purity of each subcellular fraction. HA (tagged left half) and Flag (tagged right half) were used to indicate the localization of DdCBEs. Molecular weight is given in kDa; images are representative of 2 independent biological replicates; samples are derived from the same batch of experiment and gels were processed in parallel. b, Fluorescence imaging of nuclei (DAPI, blue), HA-tagged left half (Anti-HA, red), Flag-tagged right half (Anti-Flag, green) in fixed nuclei isolated from HEK293T cells untreated or transfected with ND6-L1397N (WT) using Lipofectamine 2000, or transfected with ND6-L1397N (WT), DddA-free, UGI-free and TALE-free constructs using Lipofectamine LTX. Possible mitochondrial contamination was tested by MitoTracker (magenta). The results show that a small portion of DdCBE is localized in nuclei, regardless of the transfection conditions. TALE arrays more strongly affect the nuclear localization compared with DddA and UGI. Scale bars, 5 μm for zoomed in images of TALE-free; 40 μm for all remaining images. The images were obtained under the same exposure condition and are representative of 2 independent biological replicates.

Extended Data Fig. 6 The editing spectra of DdCBE at TAS-dependent nDNA off-target sites.

a, Sequence logos for Cs with highest Detect-seq signal obtained via WebLogo using DNA sequences at TAS-dependent off-target sites of ND6-L1397N, ND5.1-L1397N and ND4-L1397N. b, Sequence logos generated from the pTBSs of ND5.1-L1397N and ND4-L1397N. Bits reflect the level of sequence conservation at a given position. c, Aggregate distribution of C·Gs with highest Detect-seq signal across the flanking region of each pTBS for TAS-dependent off-target sites of ND6-L1397N, ND5.1-L1397N and ND4-L1397N. The position of pTBS for left or right TALE proteins is shadowed. d, A schematic illustrating the editing spectra of the three L1397N DdCBEs based on the pTBS-edits distribution analysis. Counting the first base pair after the 3’ ends of pTBS as position +1. NTD, N-terminal domain; CTD, C-terminal domain.

Extended Data Fig. 7 The TALE independency of TAS-independent off-target sites validated by targeted deep sequencing.

Results of targeted deep sequencing at five representative TAS-independent off-target sites for different ND6-L1397N constructs in Fig. 2a.

Extended Data Fig. 8 Motif search result from sequences of all TAS-independent off-target sites.

The results (with a p-value < 0.05) are generated by Tomtom program with JASPAR core motif database.

Extended Data Fig. 9 Strategies to improve the specificity of DdCBE.

a, Fusing nuclear export signal (NES) sequences into the DdCBE constructs. The protein level of DdCBE in the nucleus should be decreased, and hence lower the risk of nDNA off-target editing. b, Co-expressing DddIA that fused with nuclear localization signals (NLS). DddIA is a natural immunity protein of the deaminase DddA; bpNLS-linked DddIA is supposed to antagonize the deamination activity of DdCBEs mis-localized into the nucleus. bpNLS, bipartite NLS at both the N and C termini. c, Mutating the DddAtox in the DdCBE architecture to reduce its intrinsic DNA binding affinity. Ideally, mutated deaminase would not be able to catalyze DNA substrates without the help of simultaneously stable binding of the two TALE repeats.

Supplementary information

Supplementary Information

This file contains Supplementary Discussion, legends for Supplementary Tables 1–5 and Supplementary Videos 1 and 2, Supplementary Figures 1–27 and references.

Reporting Summary

Supplementary Table 1

Supplementary Table 2

Supplementary Table 3

Supplementary Table 4

Supplementary Table 5

Supplementary Video 1

Supplementary Video 2

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

Lei, Z., Meng, H., Liu, L. et al. Mitochondrial base editor induces substantial nuclear off-target mutations. Nature 606, 804–811 (2022). https://doi.org/10.1038/s41586-022-04836-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04836-5

This article is cited by

Comments

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

Search

Quick links

Nature Briefing: 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