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.

  • Brief Communication
  • Published:

Targeted genome editing with a DNA-dependent DNA polymerase and exogenous DNA-containing templates

Abstract

Reverse transcriptases, used in prime editing systems, exhibit lower fidelity, processivity and dNTP affinity than many DNA-dependent DNA polymerases. We report that a DNA-dependent DNA polymerase (phi29), untethered from Cas9, enables editing from a synthetic, end-stabilized DNA-containing template at up to 60% efficiency in human cells. Compared to prime editing, DNA polymerase editing avoids autoinhibitory intramolecular base pairing of the template, facilitates template synthesis and supports larger insertions (>100 nucleotides).

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: Prime editing with chimeric LPET.
Fig. 2: Phi29 DPE.

Similar content being viewed by others

Data availability

A Reporting Summary for this article is available as a supplementary information file. Plasmids for mRNA in vitro transcription, including MCP–RT, MCP–phi29 and nCas9 (H840A), have been deposited to Addgene for distribution. Illumina Sequencing data have been submitted to the Sequence Read Archive, and datasets are available under BioProject accession number PRJNA1004245 (ref. 37). Source data are provided with this paper.

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  CAS  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Zhuang, Y. et al. Increasing the efficiency and precision of prime editing with guide RNA pairs. Nat. Chem. Biol. 18, 29–37 (2022).

    Article  CAS  PubMed  Google Scholar 

  4. Wang, J. et al. Efficient targeted insertion of large DNA fragments without DNA donors. Nat. Methods 19, 331–340 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Choi, J. et al. Precise genomic deletions using paired prime editing. Nat. Biotechnol. 40, 218–226 (2022).

    Article  CAS  PubMed  Google Scholar 

  6. Jiang, T., Zhang, X.-O., Weng, Z. & Xue, W. Deletion and replacement of long genomic sequences using prime editing. Nat. Biotechnol. 40, 227–234 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. 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 

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

    Article  CAS  PubMed  Google Scholar 

  10. Liu, Y. et al. Enhancing prime editing by Csy4-mediated processing of pegRNA. Cell Res. 31, 1134–1136 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zhang, G. et al. Enhancement of prime editing via xrRNA motif-joined pegRNA. Nat. Commun. 13, 1856 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, X. et al. Enhancing prime editing efficiency by modified pegRNA with RNA G-quadruplexes. J. Mol. Cell. Biol. 14, mjac022 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Feng, Y. et al. Enhancing prime editing efficiency and flexibility with tethered and split pegRNAs. Protein Cell 14, 304–308 (2022).

    PubMed Central  Google Scholar 

  14. Ponnienselvan, K. et al. Reducing the inherent auto-inhibitory interaction within the pegRNA enhances prime editing efficiency. Nucleic Acids Res. 51, 6966–6980 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Ellefson, J. W. et al. Synthetic evolutionary origin of a proofreading reverse transcriptase. Science 352, 1590–1593 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Skasko, M. et al. Mechanistic differences in RNA-dependent DNA polymerization and fidelity between murine leukemia virus and HIV-1 reverse transcriptases. J. Biol. Chem. 280, 12190–12200 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Mathews, C. K. Deoxyribonucleotide metabolism, mutagenesis and cancer. Nat. Rev. Cancer 15, 528–539 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Wu, W.-J., Yang, W. & Tsai, M.-D. How DNA polymerases catalyse replication and repair with contrasting fidelity. Nat. Rev. Chem. 1, 0068 (2017).

    Article  CAS  Google Scholar 

  19. Truniger, V., Lázaro, J. M. & Salas, M. Two positively charged residues of phi29 DNA polymerase, conserved in protein-primed DNA polymerases, are involved in stabilisation of the incoming nucleotide. J. Mol. Biol. 335, 481–494 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Halperin, S. O. et al. CRISPR-guided DNA polymerases enable diversification of all nucleotides in a tunable window. Nature 560, 248–252 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Tou, C. J., Schaffer, D. V. & Dueber, J. E. Targeted diversification in the S. cerevisiae genome with CRISPR-guided DNA polymerase I. ACS Synth. Biol. 9, 1911–1916 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Grünewald, J. et al. Engineered CRISPR prime editors with compact, untethered reverse transcriptases. Nat. Biotechnol. 41, 337–343 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ohtsubo, Y., Sasaki, H., Nagata, Y. & Tsuda, M. Optimization of single strand DNA incorporation reaction by Moloney murine leukaemia virus reverse transcriptase. DNA Res. 25, 477–487 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Petri, K. et al. CRISPR prime editing with ribonucleoprotein complexes in zebrafish and primary human cells. Nat. Biotechnol. 40, 189–193 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. Wang, Q. et al. Broadening the reach and investigating the potential of prime editors through fully viral gene-deleted adenoviral vector delivery. Nucleic Acids Res. 49, 11986–12001 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Liu, P. et al. Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nat. Commun. 12, 2121 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Esteban, J. A., Salas, M. & Blanco, L. Fidelity of phi 29 DNA polymerase. Comparison between protein-primed initiation and DNA polymerization. J. Biol. Chem. 268, 2719–2726 (1993).

    Article  CAS  PubMed  Google Scholar 

  28. Lieberman, K. R. et al. Processive replication of single DNA molecules in a nanopore catalyzed by phi29 DNA polymerase. J. Am. Chem. Soc. 132, 17961–17972 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Anzalone, A. V. et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol. 40, 731–740 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Blanco, L. et al. Highly efficient DNA synthesis by the phage phi 29 DNA polymerase. Symmetrical mode of DNA replication. J. Biol. Chem. 264, 8935–8940 (1989).

    Article  CAS  PubMed  Google Scholar 

  31. Gillmore, J. D. et al. CRISPR–Cas9 in vivo gene editing for transthyretin amyloidosis. N. Engl. J. Med. 385, 493–502 (2021).

    Article  CAS  PubMed  Google Scholar 

  32. Lee, R. G. et al. Efficacy and safety of an investigational single-course CRISPR base-editing therapy targeting PCSK9 in nonhuman primate and mouse models. Circulation 147, 242–253 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Hopfner, K.-P. & Hornung, V. Molecular mechanisms and cellular functions of cGAS–STING signalling. Nat. Rev. Mol. Cell Biol. 21, 501–521 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Jiang, T. et al. Chemical modifications of adenine base editor mRNA and guide RNA expand its application scope. Nat. Commun. 11, 1979 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. 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  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mir, A. et al. Heavily and fully modified RNAs guide efficient SpyCas9-mediated genome editing. Nat. Commun. 9, 2641 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Liu, B. et al. Targeted genome editing with a DNA-dependent DNA polymerase and exogenous DNA-containing templates. NCBI Bioproject https://www.ncbi.nlm.nih.gov/bioproject/1004245 (2023).

Download references

Acknowledgements

We thank S. Wolfe, B. Kelch, S. Liang, P. Liu and members of the laboratories of W.X. and E.J.S. for helpful discussions as well as T. Yu and Z. Weng for help with data analysis. We also thank Ben Kleinstiver for communicating results before publication. W.X. was supported by grants from the National Institutes of Health (DP2HL137167, P01HL158506 and UH3HL147367) and the Cystic Fibrosis Foundation. X.D. and E.J.S. acknowledge support from the Leducq Foundation Transatlantic Network of Excellence Program.

Author information

Authors and Affiliations

Authors

Contributions

B.L., X.D., W.X. and E.J.S. conceptualized the project and designed experiments. B.L., X.D., C.Z. and Z.C. conducted molecular biological experiments. D.K. synthesized and purified LPETs and assisted in their design. B.L., X.D., C.Z. and H.C. conducted high-throughput sequencing and bioinformatic analyses. B.L., X.D., J.K.W., W.X. and E.J.S. interpreted the data and wrote the paper, and all authors edited the paper.

Corresponding authors

Correspondence to Wen Xue or Erik J. Sontheimer.

Ethics declarations

Competing interests

E.J.S. is a co-founder and Scientific Advisory Board member of Intellia Therapeutics and a Scientific Advisory Board member at Tessera Therapeutics. The University of Massachusetts Chan Medical School has filed patent applications related to this work. All other authors have no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers 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 LPET-mediated precision editing.

a, The levels of LPET and in vitro transcribed epegRNA were measured by qPCR after electroporation in 293T cells (n = 3). b, Diagram of LPET, MS2-less LPET, ssDNA, modified petRNA (LPET, −17), and unmodified petRNA. c, LPET and no-RT controls. HEK293T cells were electroporated with indicated mRNA (1 μg), sgRNA (100 pmol), nicking sgRNA (100 pmol), and FANCF LPET(+2), PRNP LPET (+0), IDS LPET (+0), RUNX1 LPET (+2), HBB LPET (+2). Editing was measured by deep sequencing (n = 3). Data and error bars indicate mean and s.d. of three independent biological replicates. d, Analysis of precise editing, imprecise editing, and scaffold incorporation events (FANCF) by CRISPResso2-prime editing mode (n = 3). 'MODIFIED’ represents reads containing unexpected insertions, deletions, or substitutions. Data and error bars indicate mean and s.d. of three independent biological replicates. e, Representative reads for FANCF and PRNP. Some deletions at FANCF are likely caused by short homology (underlined) near the nicking sites. Top five reads are shown.

Extended Data Fig. 2 MMLV RT-mediated precision editing with different LPET modifications.

a, Various modifications in the LPET. Precision editing by LPET with additional chemically modified residues [including Locked Nucleic Acid (LNA), 2′-F, 2′-O-methyl (OMe), 2′-fluoro (F) and 3′ phosphorothioate (PS)] as measured by deep sequencing in 293T cells (n = 3). * = 3′ PS linker. All sequences are written from 5′ to 3′. Data and error bars indicate the mean and s.d. of three independent biological replicates. b, RT mediates precise editing by LPETs with an all-DNA RTT and PBS with no modifications, or with the indicated 3′-terminal modifications, as measured by deep sequencing (n = 3). Blue and purple letters denote DNA, RNA and 2′-O-methyl RNA, respectively. Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 3 Phi29 with unmodified and modified DPETs and additional controls.

a, Precise editing mediated by Phi29. HEK293T cells were electroporated with indicated mRNA (1 μg), sgRNA (100 pmol), nicking sgRNA (100 pmol), and indicated DPET(+0), MS2-less DPET, MS2-less ssDNA (RTT+PBS), modified linear DPET(−17)), or un-modified (all-RNA) DPET. b, Precise editing mediated by Phi29 and pegRNA. Editing efficiency was measured by deep sequencing (n = 3). Data and error bars indicate the mean and s.d. of three independent biological replicates; two-tailed unpaired Student’s t-test; **P < 0.01.

Extended Data Fig. 4 Phi29 DPE with different DPET modifications.

a, Phi29-mediated precision editing with various modifications in the DPET (including locked nucleic acid, 2′-fluoro, 2′-O-methyl, and 3′ phosphorothioate) as measured by deep sequencing (n = 3). * = 3′ PS linker. All sequences are written from 5′ to 3′. Data and error bars indicate the mean and s.d. of three independent biological replicates. b, Phi29 mediates precise editing by DPETs with an all-DNA RTT and PBS with no modifications, or with the indicated 3′-terminal modifications, as measured by deep sequencing (n = 3). Blue and purple letters denote DNA, RNA and 2′-O-methyl RNA residues, respectively. Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 5 Incremental DNA replacement in the PBS/RTT at the PRNP site.

Precision editing with DNA replacement in the PBS/RTT with LPET/RT or DPET/Phi29. Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 6 LPETs and DPETs enable precise genome editing without an additional nicking sgRNA in multiple endogenous sites.

a–c, HEK293T cells were electroporated with indicated mRNAs (1 μg), pegRNA (100 pmol, PE2) and nicking sgRNA (100 pmol, PE3). LPET and DPET groups include mRNAs, sgRNA and nicking sgRNA (100 pmol), and LPET/DPET (FANCF +2 (a), PRNP +0 (b), RUNX1 +2 (c)). Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 7 LPETs and DPETs enable precise genome editing.

a–c, LPET/RT and DPET/Phi29 editing in multiple cell lines, as measured by deep sequencing (n = 3). ND, not determined. ‘SNP’ indicates the existence of a SNP in the HBB target region in the U2OS cell line, precluding comparative testing. d,e, Representative FACS plots in Fig. 2f. f, Diagram depicting mSA-RT and Phi29. The 5′-biotinylated LPET or DPET is recruited by mSA and anneals to the nicked DNA. g, Tests of biotinylated-LPETs and biotinylated-DPETs in an mCherry reporter line and at endogenous sites (n = 3). Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 8 PoII5M DNA polymerase, MarathonRT and TGIRT editing with synthetic templates.

a, Diagram depicting PoII5M/DPET editing (left). MCP-tethered PoII5M binds to the DPET via MS2. The DPET anneals to the nicked DNA strand and serves as the template for PoII5M DNA polymerase. Data indicate precision editing (3-nt substitution) at the FANCF locus with DPET (+2) and fully 2′-O-methyl (OMe)-substituted PBS (right). b, Precise editing at the FANCF locus with MarathonRT and TGIRT. Editing efficiency was measured by deep sequencing on day 3 (n = 3). Data and error bars indicate the mean and s.d. of three independent biological replicates.

Extended Data Fig. 9 Off-target and cytotoxicity evaluation.

a, On-target and off-target editing efficiencies for FANCF. Data indicate the mean and standard deviation of three independent biological replicates. b, HEK293T cells were electroporated with indicated mRNA reagents or buffers. Cell viability was measured by the CellTiter-Glo assay (n = 3) on the indicated time points. Data and error bars indicate mean and s.d. of three independent biological replicates. Two-tailed unpaired Student’s t-test: *, P < 0.05.

Extended Data Fig. 10 Precise editing for a 40-bp replacement and a 132-bp insertion.

a, Precise editing for a 40-bp insertion and 90-bp deletion at two AAVS sites (1615 and 1705) using LPET/DPET through mRNA nucleofection, followed by PCR amplification and agarose gel electrophoresis. b, Quantification of 40-bp insertion in a by deep sequencing (n = 3). Data and error bars indicate the mean and s.d. of three independent biological replicates, unless otherwise indicated. c, Diagram of the TwinPE by LPETs/DPETs for long insertion. d, Precise editing for a 132-bp insertion at the HEK3 site. Editing efficiency was measured by ddPCR (n = 2).

Source data

Supplementary information

Supplementary Information

Supplementary Table 1. Synthetic sgRNAs, nicking sgRNAs and pegRNAs. Supplementary Table 2. Synthetic LPETs and DPETs. Supplementary Table 3. Primers used for high-throughput sequencing. Supplementary Table 4. Primers used for ddPCR. Supplementary Table 5. Primers used for qPCR. Supplementary sequences.

Reporting Summary

Source Data Extended Data Fig. 10

Unmodified gel

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

Liu, B., Dong, X., Zheng, C. et al. Targeted genome editing with a DNA-dependent DNA polymerase and exogenous DNA-containing templates. Nat Biotechnol (2023). https://doi.org/10.1038/s41587-023-01947-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-023-01947-w

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing