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:

Prime editing using CRISPR-Cas12a and circular RNAs in human cells

An Author Correction to this article was published on 08 February 2024

This article has been updated

Abstract

Genome editing with prime editors based on CRISPR-Cas9 is limited by the large size of the system and the requirement for a G/C-rich protospacer-adjacent motif (PAM) sequence. Here, we use the smaller Cas12a protein to develop four circular RNA-mediated prime editor (CPE) systems: nickase-dependent CPE (niCPE), nuclease-dependent CPE (nuCPE), split nickase-dependent CPE (sniCPE) and split nuclease-dependent CPE (snuCPE). CPE systems preferentially recognize T-rich genomic regions and possess a potential multiplexing capacity in comparison to corresponding Cas9-based systems. The efficiencies of the nuclease-based systems are up to 10.42%, whereas niCPE and sniCPE reach editing frequencies of up to 24.89% and 40.75% without positive selection in human cells, respectively. A derivative system, called one-sniCPE, combines all three RNA editing components under a single promoter. By arraying CRISPR RNAs for different targets in one circular RNA, we also demonstrate low-efficiency editing of up to four genes simultaneously with the nickase prime editors niCPE and sniCPE.

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: Comparing niCPEs with prime editors with pegRNAs.
Fig. 2: CRISPR-Cas12a prime editing with niCPE, nuCPE, sniCPE and snuCPE.
Fig. 3: Expressing crRNA, circular RNA or nicking crRNA simultaneously in one-CPEs.
Fig. 4: Prime editing of multiple genes with niCPEs and sniCPEs.
Fig. 5: Effect of CPEs on off-target prime editing.

Similar content being viewed by others

Data availability

All data supporting the findings of the present study are available in the article, extended data and supplementary figures and tables, or are available from the corresponding author on request. The deep sequencing data have been deposited in an NCBI BioProject database (accession no. PRJNA1048095)42.

Change history

References

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chen, P. J. & Liu, D. R. Prime editing for precise and highly versatile genome manipulation. Nat. Rev. Genet. 24, 161–177 (2023).

    Article  CAS  PubMed  Google Scholar 

  3. Eid, A. & Qi, Y. Prime editor integrase systems boost targeted DNA insertion and beyond. Trends Biotechnol. 40, 907–909 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Gao, C. Genome engineering for crop improvement and future agriculture. Cell 184, 1621–1635 (2021).

    Article  CAS  PubMed  Google Scholar 

  5. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 40, 402–410 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Liu, B. et al. A split prime editor with untethered reverse transcriptase and circular RNA template. Nat. Biotechnol. 40, 1388–1393 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Feng, Y. et al. Enhancing prime editing efficiency and flexibility with tethered and split pegRNAs. Protein Cell 14, 304–308 (2023).

    PubMed  Google Scholar 

  9. Litke, J. L. & Jaffrey, S. R. Highly efficient expression of circular RNA aptamers in cells using autocatalytic transcripts. Nat. Biotechnol. 37, 667–675 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu, C. X. & Chen, L. L. Circular RNAs: characterization, cellular roles, and applications. Cell 185, 2016–2034 (2022).

    Article  CAS  PubMed  Google Scholar 

  11. Chen, P. J. et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell 184, 5635–5652 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lin, Q. et al. Prime genome editing in rice and wheat. Nat. Biotechnol. 38, 582–585 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Lin, Q. et al. High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat. Biotechnol. 39, 923–927 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Zong, Y. et al. An engineered prime editor with enhanced editing efficiency in plants. Nat. Biotechnol. 40, 1394–1402 (2022).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hirano, S. et al. Structure of the OMEGA nickase IsrB in complex with ωRNA and target DNA. Nature 610, 575–581 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Karvelis, T. et al. Transposon-associated TnpB is a programmable RNA-guided DNA endonuclease. Nature 599, 692–696 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yan, W. X. et al. Functionally diverse type V CRISPR-Cas systems. Science 363, 88–91 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Kim, D. et al. Genome-wide analysis reveals specificities of Cpf1 endonucleases in human cells. Nat. Biotechnol. 34, 863–868 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Zetsche, B. et al. Multiplex gene editing by CRISPR-Cpf1 using a single crRNA array. Nat. Biotechnol. 35, 31–34 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Yamano, T. et al. Crystal structure of Cpf1 in complex with guide RNA and target DNA. Cell 165, 949–962 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Merker, L., Schindele, P., Huang, T. K., Wolter, F. & Puchta, H. Enhancing in planta gene targeting efficiencies in Arabidopsis using temperature-tolerant CRISPR/LbCas12a. Plant Biotechnol. J. 18, 2382–2384 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  25. Fonfara, I., Richter, H., Bratovič, M., Le Rhun, A. & Charpentier, E. The CRISPR-associated DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA. Nature 532, 517–521 (2016).

    Article  ADS  CAS  PubMed  Google Scholar 

  26. Grünewald, J. et al. Engineered CRISPR prime editors with compact, untethered reverse transcriptases. Nat. Biotechnol. 41, 337–343 (2023).

    Article  PubMed  Google Scholar 

  27. Wu, Z., Yang, H. & Colosi, P. Effect of genome size on AAV vector packaging. Mol. Ther. 18, 80–86 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Gao, Z. et al. A truncated reverse transcriptase enhances prime editing by split AAV vectors. Mol. Ther. 30, 2942–2951 (2022).

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zheng, C. et al. A flexible split prime editor using truncated reverse transcriptase improves dual-AAV delivery in mouse liver. Mol. Ther. 30, 1343–1351 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Davis, J. R. et al. Efficient prime editing in mouse brain, liver and heart with dual AAVs. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01758-z (2023).

  31. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li, X. et al. Development of a versatile nuclease prime editor with upgraded precision. Nat. Commun. 14, 305 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Peterka, M. et al. Harnessing DSB repair to promote efficient homology-dependent and -independent prime editing. Nat. Commun. 13, 1240 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kim, Y. B. et al. A novel mechanistic framework for precise sequence replacement using reverse transcriptase and diverse CRISPR-Cas systems. Preprint at bioRxiv https://doi.org/10.1101/2022.12.13.520319 (2022).

  35. Campa, C. C., Weisbach, N. R., Santinha, A. J., Incarnato, D. & Platt, R. J. Multiplexed genome engineering by Cas12a and CRISPR arrays encoded on single transcripts. Nat. Methods 16, 887–893 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Breinig, M. et al. Multiplexed orthogonal genome editing and transcriptional activation by Cas12a. Nat. Methods 16, 51–54 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. 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 

  38. O’Sullivan, J. W. et al. Polygenic risk scores for cardiovascular disease: a scientific statement from the American Heart Association. Circulation 146, e93–e118 (2022).

    Article  MathSciNet  PubMed  PubMed Central  Google Scholar 

  39. Bischoff, N., Wimberger, S., Maresca, M. & Brakebusch, C. Improving precise CRISPR genome editing by small molecules: is there a magic potion? Cells 9, 1318 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lee, J. et al. Prime editing with genuine Cas9 nickases minimizes unwanted indels. Nat. Commun. 14, 1786 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Liang, R. et al. Prime editing using CRISPR/Cas12a and circular RNAs in human cells. National Center for Biotechnology Information (NCBI). Available at: https://www.ncbi.nlm.nih.gov/bioproject/?term=PRJNA1048095 (2023).

Download references

Acknowledgements

This work was supported by the STI 2030-Major Projects (2023ZD04074), the National Key Research and Development Program (2023YFD1202904, 2023YFF1001601 and 2023YFF1001602), and the New Cornerstone Science Foundation.

Author information

Authors and Affiliations

Authors

Contributions

C.G. and K.T.Z. designed the experiments and supervised the project. R.L., Z.H. and M.L. performed experiments. J.H., G.L., Q.G. and R.Z. performed the MiSeq data analysis. C.G., R.L., Z.H., H.Z. and J-L.Q. wrote the manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Caixia Gao.

Ethics declarations

Competing interests

The authors have submitted one patent application based on the results reported in this article. K.T.Z. is a founder and employee of Qi Biodesign. Q.G. is an employee of Qi Biodesign.

Peer review

Peer review information

Nature Biotechnology thanks Daesik Kim 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 Comparison of the editing efficiencies of Cas12a and Cas12a-D156R.

a, b, Editing efficiencies of Cas12a and Cas12a-D156R at the DNMT1-T3 site (a) and RUNX1-T4 site (b) in HEK293T cells. Frequencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in a, b.

Extended Data Fig. 2 The editing efficiencies of various prime editors.

a, Efficiencies of prime editing of various prime editors with Cas9 or nCas9 (H840A) and linear RNA (LinRNA) or circular RNA (CircRNA) at the FANCF site in HEK293T cells. b, Efficiencies of prime editing of these prime editors with Cas12a (D156R) or nCas12a (D156R-R1138A) and LinRNA or CircRNA at the DNMT1-T3 site. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in a, b. RTΔR, M-MLV RTΔRNase H.

Extended Data Fig. 3 Visualization of editing by various prime editors using the GFP reporter system.

Fields of cells labeled GFP (a), untreated sample (b), and only reporter (c) are controls. do, Microscope views of reconstitution of GFP by 12 prime editors. Scale bars, 200 µm. One of three independent experiments is shown (n = 3) in a-o. rCas12a, D156R-H759A-LbCas12a; nCas12a, D156R-R1138A-LbCas12a; rnCas12a, D156R-H759A-R1138A-LbCas12a; CircRNA, circular RNA; RTΔR, M-MLV RTΔRNase H.

Extended Data Fig. 4 Editing efficiencies of CPEs containing 5′ MS2-CircRNA, 3′ MS2-CircRNA, and 5′+3′ MS2-CircRNA.

ac, Schematic diagram of 5′ MS2-CircRTT-PBS (a), 3′ MS2-CircRTT-PBS (b), and 5′+3′ MS2-CircRTT-PBS (c). df, Schematic diagram of CPEs using 5′ MS2-CircRTT-PBS (d), 3′ MS2-CircRTT-PBS (e), and 5′+3′ MS2-CircRTT-PBS (f). g, Comparison of the editing efficiencies of CPEs containing 5′ MS2-CircRTT-PBS, 3′ MS2-CircRTT-PBS, and 5′+3′ MS2-CircRTT-PBS at different sites in HEK293T cells. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in g. Del, deletion; Sub, substitution; CircRNA, circular RNA.

Extended Data Fig. 5 Frequencies of InDels induced by CPEs.

a, The frequencies of InDel byproducts produced by CPEs at the four sites of Fig. 2i. b, The frequencies of InDel byproducts produced by CPEs at the eight sites in Fig. 2j. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in a, b. InDel editing includes imprecise prime editing byproducts.

Extended Data Fig. 6 Analysis of InDels induced by CPEs.

a, Analysis of InDels induced by CPEs at the DNMT1-T1 site in HEK293T cells. b, Analysis of InDels induced by one-CPEs at the HBB-T1 site in HeLa cells. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in a, b.

Extended Data Fig. 7 Precise editing using twin CPE2.

The editing efficiencies of CPE2-Forward, CPE2-Reverse, CPE2-two CircRNA and CPE2-one CircRNA at the SITE7 site. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3).

Extended Data Fig. 8 Comparison of the editing efficiencies of CPE2-5.

a, The editing efficiencies of niCPE2-5 and sniCPE2-5 at DNMT1-T1, DNMT3B, and HDAC1 sites. b, The editing efficiencies of one-niCPE2-5 and one-sniCPE2-5 at DNMT1-T1, HBB-T1, and HDAC1 sites. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in a, b.

Extended Data Fig. 9 Comparison of the editing efficiencies of Cas12a-CPE2 and Cas9-PE2.

The editing efficiencies of Cas9-PE2, niCPE2, sniCPE2, one-niCPE2, and one-sniCPE2. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3).

Extended Data Fig. 10 Design and delivery sniCPE2 with dual AAVs.

a, Design and packaging dual AAV vectors of sniCPE2. b, Schematic diagram of AAV delivery. c, The editing efficiencies of HEK2 and HBB-T1 double prime editings with dual AAV delivery of sniCPE2. Efficiencies (mean ± s.d.) were calculated from three independent experiments (n = 3) in c.

Supplementary information

Supplementary Information

Supplementary Figures 1–5.

Reporting Summary

Supplementary Tables

Supplementary Table1. Target sites, RT templates, PBS sequnences, desired edits and nicking crRNAs in this study. Supplementary Table 2. Sequences used for vector construction. Supplementary Table 3. MiSeq 1st PCR primers used for target sites in this study. Supplementary Table 4. MiSeq 2nd PCR primers used in this study. Supplementary Table 5. Summary of crRNA spacer sequence-like off-target sites identified by Cas-OFFinder. Supplementary Table 6. MiSeq 1st PCR primers used for off-target sites in this study.

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

Liang, R., He, Z., Zhao, K.T. et al. Prime editing using CRISPR-Cas12a and circular RNAs in human cells. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-023-02095-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-023-02095-x

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