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:

Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing

Abstract

Cytosine base editors (CBEs) efficiently generate precise C·G-to-T·A base conversions, but the activation-induced cytidine deaminase/apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (AID/APOBEC) protein family deaminase component induces considerable off-target effects and indels. To explore unnatural cytosine deaminases, we repurpose the adenine deaminase TadA-8e for cytosine conversion. The introduction of an N46L variant in TadA-8e eliminates its adenine deaminase activity and results in a TadA-8e-derived C-to-G base editor (Td-CGBE) capable of highly efficient and precise C·G-to-G·C editing. Through fusion with uracil glycosylase inhibitors and further introduction of additional variants, a series of Td-CBEs was obtained either with a high activity similar to that of BE4max or with higher precision compared to other reported accurate CBEs. Td-CGBE/Td-CBEs show very low indel effects and a background level of Cas9-dependent or Cas9-independent DNA/RNA off-target editing. Moreover, Td-CGBE/Td-CBEs are more efficient in generating accurate edits in homopolymeric cytosine sites in cells or mouse embryos, suggesting their accuracy and safety for gene therapy and other applications.

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: Engineering of TadA-8e-derived CGBEs.
Fig. 2: Evolution and characterization of Td-CBEs in mammalian cells.
Fig. 3: Off-target assessment of Td-CGBE and Td-CBEs.
Fig. 4: Examination of mouse embryos with Td-CGBE.
Fig. 5: Precise editing of pathogenic SNVs by TadA-derived base editors and target library analysis for eTd-CBEs.

Similar content being viewed by others

Data availability

HTS data have been deposited in the NCBI Sequence Read Archive database under accession codes PRJNA822038, PRJNA871961, PRJNA855334, PRJNA835691, PRJNA835701 and PRJNA882574 (refs. 47,48,49,50,51,52). RNA-seq data have been deposited in the NCBI Sequence Read Archive database under accession codes PRJNA871962 and PRJNA830998 (refs. 53,54). There are no restrictions on data availability. Source data are provided with this paper.

Code availability

The relevant codes of analysis for Detect-seq data were deposited in GitHub (https://github.com/menghaowei/Detect-seq)55.

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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kurt, I. C. et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells. Nat. Biotechnol. 39, 41–46 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Zhao, D. et al. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat. Biotechnol. 39, 35–40 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Koblan, L. W. et al. Efficient C·G-to-G·C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nat. Biotechnol. 39, 1414–1425 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen, L. et al. Programmable C:G to G:C genome editing with CRISPR-Cas9-directed base excision repair proteins. Nat. Commun. 12, 1384 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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  PubMed  PubMed Central  Google Scholar 

  15. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chen, L. et al. Engineering precise adenine base editor with infinitesimal rates of bystander mutations and off-target editing. Nat. Chem. Biol. https://doi.org/10.1038/s41589-022-01163-8 (2022).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jeong, Y. K. et al. Adenine base editor engineering reduces editing of bystander cytosines. Nat. Biotechnol. 39, 1426–1433 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Tan, J., Zhang, F., Karcher, D. & Bock, R. Engineering of high-precision base editors for site-specific single nucleotide replacement. Nat. Commun. 10, 439 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Thuronyi, B. W. et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat. Biotechnol. 37, 1070–1079 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Nishimasu, H. et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 361, 1259–1262 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zhang, X. et al. Increasing the efficiency and targeting range of cytidine base editors through fusion of a single-stranded DNA-binding protein domain. Nat. Cell Biol. 22, 740–750 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. Lee, S. et al. Single C-to-T substitution using engineered APOBEC3G-nCas9 base editors with minimum genome- and transcriptome-wide off-target effects. Sci. Adv. 6, eaba1773 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

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

    Article  PubMed  Google Scholar 

  28. 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 

  29. Lei, Z. et al. Mitochondrial base editor induces substantial nuclear off-target mutations. Nature 606, 804–811 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Yuan, T. et al. Optimization of C-to-G base editors with sequence context preference predictable by machine learning methods. Nat. Commun. 12, 4902 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Auer-Grumbach, M., Strasser-Fuchs, S., Robl, T., Windpassinger, C. & Wagner, K. Late onset Charcot-Marie-Tooth 2 syndrome caused by two novel mutations in the MPZ gene. Neurology 61, 1435–1437 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Rodriguez-Escudero, I. et al. A comprehensive functional analysis of PTEN mutations: implications in tumor- and autism-related syndromes. Hum. Mol. Genet. 20, 4132–4142 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Syrbe, S. et al. De novo loss- or gain-of-function mutations in KCNA2 cause epileptic encephalopathy. Nat. Genet. 47, 393–399 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Esteghamat, F. et al. CELA2A mutations predispose to early-onset atherosclerosis and metabolic syndrome and affect plasma insulin and platelet activation. Nat. Genet. 51, 1233–1243 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ropero, P. et al. Hb Johnstown [β 109 (G11) Val–>Leu]: second case described and associated for the first time with β0-thalassemia in two Spanish families. Am. J. Hematol. 65, 298–301 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Fazeli, W. et al. A TUBB6 mutation is associated with autosomal dominant non-progressive congenital facial palsy, bilateral ptosis and velopharyngeal dysfunction. Hum. Mol. Genet. 26, 4055–4066 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Wu, C. H. et al. Mutations in the profilin 1 gene cause familial amyotrophic lateral sclerosis. Nature 488, 499–503 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Arbab, M. et al. Determinants of base editing outcomes from target library analysis and machine learning. Cell 182, 463–480 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Walton, R. T., Christie, K. A., Whittaker, M. N. & Kleinstiver, B. P. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 368, 290–296 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Li, J. et al. Structure-guided engineering of adenine base editor with minimized RNA off-targeting activity. Nat. Commun. 12, 2287 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Shi, K. et al. Structural basis for targeted DNA cytosine deamination and mutagenesis by APOBEC3A and APOBEC3B. Nat. Struct. Mol. Biol. 24, 131–139 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Zhang, X. et al. Dual base editor catalyzes both cytosine and adenine base conversions in human cells. Nat. Biotechnol. 38, 856–860 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Kruger, S. et al. Sensorimotor polyneuropathy and systemic amyloidosis as paraneoplastic symptoms of a carcinoid-like well differentiated carcinoma of the breast. Dtsch. Med. Wochenschr. 123, 179–184 (1998).

    CAS  PubMed  Google Scholar 

  45. Li, D. et al. Heritable gene targeting in the mouse and rat using a CRISPR-Cas system. Nat. Biotechnol. 31, 681–683 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA822038 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA822038 (2022).

  48. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA871961 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA871961 (2022).

  49. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA855334 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA855334 (2022).

  50. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA835691 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA835691 (2022).

  51. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA835701 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA835701 (2022).

  52. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA882574 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA882574 (2022).

  53. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA871962 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA871962 (2022).

  54. Chen, L. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. NCBI SRA, BioProject PRJNA830998 https://www.ncbi.nlm.nih.gov/bioproject/PRJNA830998 (2022).

  55. Chen, L. et al. Td-BE-Detect-seq analysis. GitHub https://github.com/menghaowei/Detect-seq (2022).

Download references

Acknowledgements

We are grateful to the East China Normal University Public Platform for Innovation (011). We thank Y. Zhang from the Flow Cytometry Core Facility of the School of Life Sciences in ECNU and H. Jiang from the Core Facility and Technical Service Center for the SLSB of the School of Life Sciences and Biotechnology in SJTU. We thank L. Ji (MedSci) for designing schematic diagrams. This work is partially supported by grants from the National Key R&D Program of China (2019YFA0802800 to M.L., 2019YFA0110802 to D.L., 2019YFA0802200 to C.Y., and 2019YFA0110900 to C.Y.), the National Natural Science Foundation of China (32025023 to D.L., 32230064 to D.L., 31971366 to L.W., 82230002 to M.L., 21825701 to C.Y., 91953201 to C.Y. and 92153303 to C.Y.), the Shanghai Municipal Commission for Science and Technology (21CJ1402200 to D.L. and 20140900200 to D.L.), and the Innovation Program of the Shanghai Municipal Education Commission (2019-01-07-00-05-E00054 to D.L.), the Fundamental Research Funds for the Central Universities (NK2022010207 to D.L.), the State Key Laboratory of Drug Research (SIMM2205KF-01 to C.Y.) and support from the East China Normal University Outstanding Doctoral Students Academic Innovation Ability Improvement Project (YBNLTS2021-026 to L.C.).

Author information

Authors and Affiliations

Authors

Contributions

L.C. and D.L. designed the experiments. L.C., B.Z., G.R., H.M., Y.Y., M.H., C.L., S.Z., H.G., S.B., C.L., R.D. and N.X. performed the experiments. L.C., B.Z., G.R., H.M., Y.Y., M.H., D.Z., C.L., H.W., C.L., Z.L., Y.C., Y.G., S.S., C.Y., G.S., L.W., C.Y., M.L. and D.L. analyzed the data. D.L., L.C., B.Z., G.R., H.M., C.Y. and G.S. wrote the manuscript with inputs from all the authors. D.L. supervised the research.

Corresponding authors

Correspondence to Chengqi Yi, Mingyao Liu or Dali Li.

Ethics declarations

Competing interests

The authors have submitted patent applications based on the results reported in this study (L.C., D.L., G.R., C.L., H.G., B.Z., J.Y., S.B., R.D. and M.L.). The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Francisco Sanchez-Rivera 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–18, Supplementary Tables 1–4, Supplementary Sequences 1–3 and Supplementary Note

Reporting Summary

Supplementary Tables

Supplementary Tables 5 and 6

Supplementary Data

Source data for Supplementary Figs. 1–6, 8–12, 16 and 17.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source.

Source Data Fig. 5

Statistical source data.

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

Chen, L., Zhu, B., Ru, G. et al. Re-engineering the adenine deaminase TadA-8e for efficient and specific CRISPR-based cytosine base editing. Nat Biotechnol 41, 663–672 (2023). https://doi.org/10.1038/s41587-022-01532-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01532-7

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