Glycosylase base editors enable C-to-A and C-to-G base changes (2024)

  • Letter
  • Published:

Nature Biotechnology volume39,pages 35–40 (2021)Cite this article

  • 21k Accesses

  • 262 Citations

  • 68 Altmetric

  • Metrics details

Subjects

  • Biotechnology
  • Molecular biology

A Publisher Correction to this article was published on 29 July 2020

This article has been updated

Abstract

Current base editors (BEs) catalyze only base transitions (C to T and A to G) and cannot produce base transversions. Here we present BEs that cause C-to-A transversions in Escherichia coli and C-to-G transversions in mammalian cells. These glycosylase base editors (GBEs) consist of a Cas9 nickase, a cytidine deaminase and a uracil-DNA glycosylase (Ung). Ung excises the U base created by the deaminase, forming an apurinic/apyrimidinic (AP) site that initiates the DNA repair process. In E. coli, we used activation-induced cytidine deaminase (AID) to construct AID-nCas9-Ung and found that it converts C to A with an average editing specificity of 93.8% ± 4.8% and editing efficiency of 87.2% ± 6.9%. For use in mammalian cells, we replaced AID with rat APOBEC1 (APOBEC-nCas9-Ung). We tested APOBEC-nCas9-Ung at 30 endogenous sites, and we observed C-to-G conversions with a high editing specificity at the sixth position of the protospacer between 29.7% and 92.2% and an editing efficiency between 5.3% and 53.0%. APOBEC-nCas9-Ung supplements the current adenine and cytidine BEs (ABE and CBE, respectively) and could be used to target G/C disease-causing mutations.

This is a preview of subscription content, access via your institution

Access options

Access through your institution

Change institution

Buy or subscribe

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

$29.99 /30days

cancel any time

Learn more

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Learn more

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Glycosylase base editors enable C-to-A and C-to-G base changes (1)
Glycosylase base editors enable C-to-A and C-to-G base changes (2)
Glycosylase base editors enable C-to-A and C-to-G base changes (3)
Glycosylase base editors enable C-to-A and C-to-G base changes (4)

Similar content being viewed by others

Glycosylase base editors enable C-to-A and C-to-G base changes (6)

Plasmid targeting and destruction by the DdmDE bacterial defence system

Article 13 May 2024

Glycosylase base editors enable C-to-A and C-to-G base changes (7)

Improving prime editing with an endogenous small RNA-binding protein

Article Open access 03 April 2024

Data availability

There is no restriction on data associated with this study. The raw data for Figs. 1 and 4 and Extended Data Fig. 1 have been submitted with the manuscript. High-throughput sequencing data have been deposited in the NCBI database (accession code SRP265375). Source data are provided with this paper.

Change history

References

  1. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article CAS Google Scholar

  2. Fujii, W., Kawasaki, K., Sugiura, K. & Naito, K. Efficient generation of large-scale genome-modified mice using gRNA and Cas9 endonuclease. Nucleic Acids Res. 41, e187 (2013).

    Article CAS Google Scholar

  3. Hwang, W. Y. et al. Efficient genome editing in zebrafish using a CRISPR–Cas9 system. Nat. Biotechnol. 31, 227–229 (2013).

    Article CAS Google Scholar

  4. Shalem, O. et al. Genome-scale CRISPR–Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article CAS Google Scholar

  5. Jiang, Y. et al. Multigene editing in the Escherichia coli genome via the CRISPR–Cas9 system. Appl. Environ. Microbiol. 81, 2506–2514 (2015).

    Article CAS Google Scholar

  6. 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

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

  8. 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 Google Scholar

  9. Ma, Y. et al. Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells. Nat. Methods 13, 1029–1035 (2016).

    Article CAS Google Scholar

  10. Zhang, Y. et al. Programmable base editing of zebrafish genome using a modified CRISPR–Cas9 system. Nat. Commun. 8, 118 (2017).

    Article CAS Google Scholar

  11. Banno, S., Nishida, K., Arazoe, T., Mitsunobu, H. & Kondo, A. Deaminase-mediated multiplex genome editing in Escherichia coli. Nat. Microbiol. 3, 423–429 (2018).

    Article CAS Google Scholar

  12. Li, C. et al. Expanded base editing in rice and wheat using a Cas9–adenosine deaminase fusion. Genome Biol. 19, 59 (2018).

    Article Google Scholar

  13. Ryu, S.-M. et al. Adenine base editing in mouse embryos and an adult mouse model of duch*enne muscular dystrophy. Nat. Biotechnol. 36, 536–539 (2018).

    Article CAS Google Scholar

  14. Shimatani, Z. et al. Inheritance of co-edited genes by CRISPR-based targeted nucleotide substitutions in rice. Plant Physiol. Biochem. 131, 78–83 (2018).

  15. Wang, Y. et al. MACBETH: multiplex automated Corynebacterium glutamicum base editing method. Metab. Eng. 47, 200–210 (2018).

    Article CAS Google Scholar

  16. Hess, G. T. et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

    Article CAS Google Scholar

  17. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168, 20–36 (2016).

    Article Google Scholar

  18. Kim, K. et al. Highly efficient RNA-guided base editing in mouse embryos. Nat. Biotechnol. 35, 435–437 (2017).

    Article CAS Google Scholar

  19. Lindahl, T. An N-glycosidase from Escherichia coli that releases free uracil from DNA containing deaminated cytosine residues. Proc. Natl Acad. Sci. USA 71, 3649–3653 (1974).

    Article CAS Google Scholar

  20. Zheng, K. et al. Highly efficient base editing in bacteria using a Cas9–cytidine deaminase fusion. Commun. Biol. 1, 32 (2018).

    Article Google Scholar

  21. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–191 (2015).

    Article CAS Google Scholar

  22. Kleinstiver, B. P. et al. Engineered CRISPR–Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    Article Google Scholar

  23. Hu, J. H. et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 556, 57–63 (2018).

    Article CAS Google Scholar

  24. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).

    Article CAS Google Scholar

  25. Engler, C., Kandzia, R. & Marillonnet, S. A one pot, one step, precision cloning method with high- throughput capability. PLoS ONE 3, e3647 (2008).

    Article Google Scholar

  26. Hillson, N. J., Rosengarten, R. D. & Keasling, J. D. j5 DNA assembly design automation software. ACS Synth. Biol. 1, 14–21 (2011).

    Article Google Scholar

  27. Zhao, J. et al. Engineering central metabolic modules of Escherichia coli for improving β-carotene production. Metab. Eng. 17, 42–50 (2013).

    Article CAS Google Scholar

  28. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article CAS Google Scholar

  29. Feng, X., Zhao, D., Zhang, X., Ding, X. & Bi, C. CRISPR/Cas9 assisted multiplex genome editing technique in Escherichia coli. Biotechnol. J. 13, e1700604 (2018).

  30. 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

Download references

Acknowledgements

This research was financially supported by the National Key Research and Development Program of China (2019YFA0904900), the Key Research Program of the Chinese Academy of Science (KFZD-SW-215), the National Natural Science Foundation of China (31861143019), a Newton Fund PhD placement program grant (ID 352639434) under the UK–China Joint Research and Innovation Partnership Fund and the Tianjin Synthetic Biotechnology Innovation Capacity Improvement Project (TSBICIP-PTJS-003). We gratefully thank F. Gu (Wenzhou Medical University) for providing the plasmid pAPOBEC-nCas9-UGI and Y. Li (Tianjin Institute of Industrial Biotechnology) for assisting with the data analysis.

Author information

Author notes

  1. These authors contributed equally: Dongdong Zhao, Ju Li, Siwei Li.

Authors and Affiliations

  1. Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China

    Dongdong Zhao,Siwei Li,Xiuqing Xin,Muzi Hu,Changhao Bi&Xueli Zhang

  2. College of Life Science, Tianjin Normal University, Tianjin, China

    Ju Li

  3. College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China

    Xiuqing Xin

  4. School of Biological Engineering, Dalian Polytechnic University, Dalian, China

    Muzi Hu

  5. Centre for Synthetic and Systems Biology and UK Centre for Mammalian Synthetic Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK

    Marcus A. Price&Susan J. Rosser

Authors

  1. Dongdong Zhao

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  2. Ju Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  3. Siwei Li

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  4. Xiuqing Xin

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  5. Muzi Hu

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  6. Marcus A. Price

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  7. Susan J. Rosser

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  8. Changhao Bi

    View author publications

    You can also search for this author in PubMedGoogle Scholar

  9. Xueli Zhang

    View author publications

    You can also search for this author in PubMedGoogle Scholar

Contributions

X.Z., C.B. and J.L. designed the research, analyzed data and wrote the manuscript. D.Z. designed the research, performed experiments, analyzed data and wrote the manuscript. S.L. performed experiments and analyzed data. M.A.P. performed experiments and wrote the manuscript. S.J.R. wrote the manuscript. X.X. and M.H. performed experiments.

Corresponding authors

Correspondence to Changhao Bi or Xueli Zhang.

Ethics declarations

Competing interests

A provisional patent has been submitted in part entailing the reported approach.

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 Sequencing data results following CBE.

a, Results obtained following treatment with nCas9-AID in E. coli MG1655 Δung. b, Results obtained following treatment with dCas9-AID in wild type E. coli MG1655. For all plots, dots represent individual biological replicates, bars represent mean values, and error bars represent the s.d. of three independent biological replicates.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1, 2, 4 and 7; Supplementary Figs. 1 and 2; and DNA sequences for vector components.

Supplementary Table 3

Mutations of APOBEC-nCas9-UGI and APOBEC-nCas9-Ung at RP11-177B4-3, PSMB2-1 and EMX1-site5 using mismatched sgRNAs.

Supplementary Table 5

The main primers used for this work.

Supplementary Table 6

The main plasmids used for this work.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Rights and permissions

About this article

Glycosylase base editors enable C-to-A and C-to-G base changes (8)

Cite this article

Zhao, D., Li, J., Li, S. et al. Glycosylase base editors enable C-to-A and C-to-G base changes. Nat Biotechnol 39, 35–40 (2021). https://doi.org/10.1038/s41587-020-0592-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-020-0592-2

This article is cited by

Glycosylase base editors enable C-to-A and C-to-G base changes (2024)

References

Top Articles
Latest Posts
Article information

Author: Greg O'Connell

Last Updated:

Views: 6354

Rating: 4.1 / 5 (42 voted)

Reviews: 89% of readers found this page helpful

Author information

Name: Greg O'Connell

Birthday: 1992-01-10

Address: Suite 517 2436 Jefferey Pass, Shanitaside, UT 27519

Phone: +2614651609714

Job: Education Developer

Hobby: Cooking, Gambling, Pottery, Shooting, Baseball, Singing, Snowboarding

Introduction: My name is Greg O'Connell, I am a delightful, colorful, talented, kind, lively, modern, tender person who loves writing and wants to share my knowledge and understanding with you.