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

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

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

Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature576, 149–157 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

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 

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 

Wang, J. et al. Efficient targeted insertion of large DNA fragments without DNA donors. Nat. Methods19, 331–340 (2022).

Article 
CAS 
PubMed 

Google Scholar 

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

Article 
CAS 
PubMed 

Google Scholar 

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 

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 

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 

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

Article 
CAS 
PubMed 

Google Scholar 

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 

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 

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 

Feng, Y. et al. Enhancing prime editing efficiency and flexibility with tethered and split pegRNAs. Protein Cell14, 304–308 (2022).

PubMed Central 

Google Scholar 

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 

Ellefson, J. W. et al. Synthetic evolutionary origin of a proofreading reverse transcriptase. Science352, 1590–1593 (2016).

Article 
CAS 
PubMed 

Google Scholar 

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 

Mathews, C. K. Deoxyribonucleotide metabolism, mutagenesis and cancer. Nat. Rev. Cancer15, 528–539 (2015).

Article 
CAS 
PubMed 

Google Scholar 

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 

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 

Halperin, S. O. et al. CRISPR-guided DNA polymerases enable diversification of all nucleotides in a tunable window. Nature560, 248–252 (2018).

Article 
CAS 
PubMed 

Google Scholar 

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 

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 

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 

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 

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 

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 

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 

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 

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 

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 

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 

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. Circulation147, 242–253 (2023).

Article 
CAS 
PubMed 

Google Scholar 

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 

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 

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 

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 

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

Author notes

Xiaolong Dong

Present address: Tessera Therapeutics, Somerville, MA, USA

These authors contributed equally: Bin Liu, Xiaolong Dong, Chunwei Zheng.

Authors and Affiliations

RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA

Bin Liu, Xiaolong Dong, Chunwei Zheng, David Keener, Zexiang Chen, Haoyang Cheng, Jonathan K. Watts, Wen Xue & Erik J. Sontheimer

Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA

Jonathan K. Watts, Wen Xue & Erik J. Sontheimer

Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA

Jonathan K. Watts & Erik J. Sontheimer

Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA

Wen Xue

Department of Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA

Wen Xue & Erik J. Sontheimer

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
>>> Read full article>>>
Copyright for syndicated content belongs to the linked Source : Nature.com – https://www.nature.com/articles/s41587-023-01947-w

Exit mobile version