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:

Development of miniature base editors using engineered IscB nickase

Abstract

As a miniature RNA-guided endonuclease, IscB is presumed to be the ancestor of Cas9 and to share similar functions. IscB is less than half the size of Cas9 and thus more suitable for in vivo delivery. However, the poor editing efficiency of IscB in eukaryotic cells limits its in vivo applications. Here we describe the engineering of OgeuIscB and its corresponding ωRNA to develop an IscB system that is highly efficient in mammalian systems, named enIscB. By fusing enIscB with T5 exonuclease (T5E), we found enIscB-T5E exhibited comparable targeting efficiency to SpG Cas9 while showing reduced chromosome translocation effects in human cells. Furthermore, by fusing cytosine or adenosine deaminase with enIscB nickase, we generated miniature IscB-derived base editors (miBEs), exhibiting robust editing efficiency (up to 92%) to induce DNA base conversions. Overall, our work establishes enIscB-T5E and miBEs as versatile tools for genome editing.

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 IscB ωRNA and protein to improve editing efficiency in mammalian cells.
Fig. 2: The enIscB with T5 exonuclease enables robust genomic editing in mammalian cells with high specificity.
Fig. 3: A-to-I and C-to-T base editors with enIscB.
Fig. 4: The enIscB nickase-derived base editors enable robust genomic editing with high specificity in mammalian cells.

Similar content being viewed by others

Data availability

All the PEM-seq data have been deposited in the National Center for Biotechnology Information Sequence Read Archive under project accession number PRJNA889595 and the Sequence Read Archive numbers are provided in Supplementary Table 1. The related plasmids have been deposited to Addgene. All materials are available upon reasonable request. Source data are provided with this paper.

Code availability

Bioinformatics codes were deposited in GitHub repository (https://github.com/yszhou2016/IscB).

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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Burnett, C. A. et al. Examination of the cell cycle dependence of cytosine and adenine base editors. Front. Genome Ed. 4, 923718 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Kleinstiver, B. P. et al. Engineered CRISPR-Cas12a variants with increased activities and improved targeting ranges for gene, epigenetic and base editing. Nat. Biotechnol. 37, 276–282 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Kim, D. Y. et al. Hypercompact adenine base editors based on transposase B guided by engineered RNA. Nat. Chem. Biol. 18, 1005–1013 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Xu, X. et al. Engineered miniature CRISPR-Cas system for mammalian genome regulation and editing. Mol. Cell 81, 4333–4345 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Chen, S. et al. Compact Cje3Cas9 for efficient in vivo genome editing and adenine base editing. CRISPR J. 5, 472–486 (2022).

    Article  CAS  PubMed  Google Scholar 

  10. Zhang, H. et al. Adenine base editing in vivo with a single adeno-associated virus vector. GEN Biotechnol. 1, 285–299 (2022).

    Article  CAS  PubMed  Google Scholar 

  11. Davis, J. R. et al. Efficient in vivo base editing via single adeno-associated viruses with size-optimized genomes encoding compact adenine base editors. Nat. Biomed. Eng. 6, 1272–1283 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Altae-Tran, H. et al. The widespread IS200/IS605 transposon family encodes diverse programmable RNA-guided endonucleases. Science 374, 57–65 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schuler, G., Hu, C. & Ke, A. Structural basis for RNA-guided DNA cleavage by IscB-ωRNA and mechanistic comparison with Cas9. Science 376, 1476–1481 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Zhang, H. et al. An engineered xCas12i with high activity, high specificity and broad PAM range. Protein Cell https://doi.org/10.1093/procel/pwac052 (2022).

  16. Spencer, J. M. & Zhang, X. Deep mutational scanning of S. pyogenes Cas9 reveals important functional domains. Sci. Rep. 7, 16836 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Guo, L. Y. et al. Multiplexed genome regulation in vivo with hyper-efficient Cas12a. Nat. Cell Biol. 24, 590–600 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. McGaw, C. et al. Engineered Cas12i2 is a versatile high-efficiency platform for therapeutic genome editing. Nat. Commun. 13, 2833 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wu, Y. et al. Improving FnCas12a genome editing by exonuclease fusion. CRISPR J. 3, 503–511 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Yin, J. et al. Cas9 exo-endonuclease eliminates chromosomal translocations during genome editing. Nat. Commun. 13, 1204 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Yin, J. et al. Optimizing genome editing strategy by primer-extension-mediated sequencing. Cell Disco. 5, 18 (2019).

    Article  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kechin, A., Boyarskikh, U., Kel, A. & Filipenko, M. cutPrimers: a new tool for accurate cutting of primers from reads of targeted next generation sequencing. J. Comput Biol. 24, 1138–1143 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liu, Y. et al. PEM-seq comprehensively quantifies DNA repair outcomes during gene-editing and DSB repair. Star. Protoc. 3, 101088 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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 

Download references

Acknowledgements

This work was supported by HUIDAGENE Therapeutics Inc., HUIEDIT Therapeutics Inc.

Author information

Authors and Affiliations

Authors

Contributions

Y.Z. and H.Y. jointly conceived the project. D.H. and Q.X. jointly designed experiments. D.H. and Y.W. performed ωRNA and protein engineering and endogenous sites cleavage assay. Q.X. conducted base editors-related experiments, including engineering IscB nickases, base editors efficiency comparison and off-target analysis. D.H. performed off-target assays. H.Z., W.Z., J.Z. and L.B. performed PEM-seq experiments. Y.Z. performed bioinformatics analysis. X.D., G.L., X.K., S.W., J.S., Y.Y., N.Z. and L.S. assisted with experiments. H.Y. and Y.Z. supervised the whole project. Y.Z. and H.Y. wrote the manuscript with data contributed by all authors who participated in the project.

Corresponding authors

Correspondence to Hui Yang or Yingsi Zhou.

Ethics declarations

Competing interests

Y.Z., D.H. and Q.X. have filed patent applications related to this work through HUIDAGENE and CAS. H.Y. and L.S. are cofounders of HUIDAGENE Therapeutics. H.Y. and Y.Z. are cofounders of HUIEDIT Therapeutics. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Lei Tang and Rita Strack, in collaboration with the Nature Methods team.

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 Effect of truncation of R1, R2, R3 and R4 regions of ωRNA with 5-nt.

Each point represents the editing efficiency of independent biological replicate by FACS analysis. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Extended Data Fig. 2 Structure-based engineering of IscB at ten amino acid sites.

Wild type ωRNA was used in this experiment. Each point represents the editing efficiency of independent biological replicate by FACS analysis. Data are presented as means ± s.d., n = 3. P-values determined by Sidak’s multiple comparisons test following ordinary one-way analysis of variance.

Source data

Extended Data Fig. 3 TAM profiling of IscB-ωRNA, IscB*-ωRNA, IscB-ωRNA* and IscB*-ωRNA*.

The panels from left to right are 5′-NWGGNA-3′, 5′-NWAGNA-3′, 5′-NWGANA-3′ and 5′-NWAANA-3′ TAMs profiling of IscB-ωRNA, IscB*-ωRNA, IscB-ωRNA* and IscB*-ωRNA* in HEK293T cells with the GFxxFP fluorescence reporter systems, which contain different TAMs. Colors reflect the mean of three independent biological replicates.

Source data

Extended Data Fig. 4 Comparison of cleavage efficiency of the wild-type IscB after fusing T5 exonuclease at N- and C-terminals.

IscB-T5E and T5E-IscB represent fusing T5 exonuclease at C-terminal and N-terminal respectively. IscB represents WT-IscB without T5E. NT, non-targeting gRNA. Each point represents the editing efficiency of independent biological replicate by FACS analysis. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Extended Data Fig. 5 Comparison the indel features of enIscB-T5E, enIscB and SpG.

a. The cleavage patterns of small deletions and insertions generated by enIscB-T5E, enIscB and SpG in HEK-293T cells at EMX1-sg1 site. b. Length distribution of deletions and insertions generated by enIscB-T5E, enIscB and SpG in HEK-293T cells at EMX1-sg1 site.

Source data

Extended Data Fig. 6 Off-target efficiency of enIscB, enIscB-T5E and SpG at in-silico predicted off-target sites of EMX1, ALDH1A3 and VEGFA genes.

b. off-target effects of enIscB and enIscB-T5E (a) and SpG (b) targeting ALDH1A3 gene respectively. c-d. off-target effects of enIscB and enIscB-T5E (c) and SpG (d) targeting EMX1 gene respectively. e-f. off-target effects of enIscB and enIscB-T5E (e) and SpG (f) targeting VEGFA gene respectively. The underlined sequences represent the on-target or predicted off-target sites. The mismatches were labeled in red and lower case. Each point represents the editing efficiency of independent biological replicate by deep sequencing analysis. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Extended Data Fig. 7 Base editing activity window plots showing mean A-to-G and C-to-T editing at all tested target positions.

Top represents ABEs and bottom represents CBEs. Each point represents the average editing efficiency of three independent biological replicates measured at each endogenous locus. Data are presented as means ± SEM.

Source data

Extended Data Fig. 8 Indel frequency of base editors.

Left represents ABEs and right represents CBEs. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Extended Data Fig. 9 The gRNA-dependent off-target levels of CBEs at the in-silico predicted off-target sites.

The left and right panel is off-target effects of miCBE and SpG-CBE targeting EMX1, VEGFA and PCSK9 genes respectively. The underlined sequences represent the on-target or predicted off-target sites. The mismatches were labeled in red and lower case. Each point represents the editing efficiency of independent biological replicates by deep sequencing analysis. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Extended Data Fig. 10 The gRNA-dependent off-target levels of ABEs at the in-silico predicted off-target sites.

The left and right panel is off-target effects of miABE and SpG-ABE targeting EMX1, PCSK9 and TTR genes respectively. The underlined sequences represent the on-target or predicted off-target sites. The mismatches were labeled in red and lower case. Each point represents the editing efficiency of independent biological replicate by deep sequencing analysis. Data are presented as means ± s.d., n = 3 independent biological replicates.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5.

Reporting Summary

Peer Review File

Supplementary Table 1

Primers and sequences.

Supplementary Table 2

Predicted off-target sites from Cas-OFFinder.

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 data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

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

Han, D., Xiao, Q., Wang, Y. et al. Development of miniature base editors using engineered IscB nickase. Nat Methods 20, 1029–1036 (2023). https://doi.org/10.1038/s41592-023-01898-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-01898-9

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