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:

A split prime editor with untethered reverse transcriptase and circular RNA template

Abstract

Delivery and optimization of prime editors (PEs) have been hampered by their large size and complexity. Although split versions of genome-editing tools can reduce construct size, they require special engineering to tether the binding and catalytic domains. Here we report a split PE (sPE) in which the Cas9 nickase (nCas9) remains untethered from the reverse transcriptase (RT). The sPE showed similar efficiencies in installing precise edits as the parental unsplit PE3 and no increase in insertion–deletion (indel) byproducts. Delivery of sPE to the mouse liver with hydrodynamic injection to modify β-catenin drove tumor formation with similar efficiency as PE3. Delivery with two adeno-associated virus (AAV) vectors corrected the disease-causing mutation in a mouse model of type I tyrosinemia. Similarly, prime editing guide RNAs (pegRNAs) can be split into a single guide RNA (sgRNA) and a circular RNA RT template to increase flexibility and stability. Compared to previous sPEs, ours lacks inteins, protein–protein affinity modules and nuclease-sensitive pegRNA extensions, which increase construct complexity and might reduce efficiency. Our modular system will facilitate the delivery and optimization of PEs.

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

Access options

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

Fig. 1: sPE enables genome editing in cells and in adult mouse liver.
Fig. 2: sPE dual AAV rescues weight loss in Fah-mutant mice.
Fig. 3: Prime editing by modular RNA components.
Fig. 4: Effective prime editing by sPE through mRNA and RNP nucleofection.

Similar content being viewed by others

Data availability

A Reporting Summary for this article is available as a supplementary information file. The raw DNA sequencing data are available at the NCBI Sequence Read Archive database under project number PRJNA802843. Plasmids for mammalian expression of MS2-pegRNA, petRNA, alternative PEs and split RTs as well as for bacterial expression of recombinant NLS-M-MLV RT have been deposited to Addgene for distribution. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  4. Wang, D., Zhang, F. & Gao, G. CRISPR-based therapeutic genome editing: strategies and in vivo delivery by AAV vectors. Cell 181, 136–150 (2020).

    Article  CAS  Google Scholar 

  5. Truong, D. J. et al. Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic Acids Res. 43, 6450–6458 (2015).

    Article  CAS  Google Scholar 

  6. Maji, B. et al. Multidimensional chemical control of CRISPR–Cas9. Nat. Chem. Biol. 13, 9–11 (2017).

    Article  CAS  Google Scholar 

  7. Liu, K. I. et al. A chemical-inducible CRISPR–Cas9 system for rapid control of genome editing. Nat. Chem. Biol. 12, 980–987 (2016).

    Article  CAS  Google Scholar 

  8. Li, C. et al. SWISS: multiplexed orthogonal genome editing in plants with a Cas9 nickase and engineered CRISPR RNA scaffolds. Genome Biol. 21, 141 (2020).

    Article  Google Scholar 

  9. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR–Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  Google Scholar 

  10. Wang, Y. et al. sgBE: a structure-guided design of sgRNA architecture specifies base editing window and enables simultaneous conversion of cytosine and adenosine. Genome Biol. 21, 222 (2020).

    Article  Google Scholar 

  11. Jiang, W. et al. BE-PLUS: a new base editing tool with broadened editing window and enhanced fidelity. Cell Res. 28, 855–861 (2018).

    Article  CAS  Google Scholar 

  12. Jang, H. et al. Application of prime editing to the correction of mutations and phenotypes in adult mice with liver and eye diseases. Nat. Biomed. Eng. 6, 181–194 (2021).

    Article  Google Scholar 

  13. Zhi, S. et al. Dual-AAV delivering split prime editor system for in vivo genome editing. Mol Ther. 30, 283–294 (2022).

    Article  CAS  Google Scholar 

  14. Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-01039-7 (2021).

  15. Zhao, C., Liu, F. & Pyle, A. M. An ultraprocessive, accurate reverse transcriptase encoded by a metazoan group II intron. RNA 24, 183–195 (2018).

    Article  CAS  Google Scholar 

  16. Mohr, S. et al. Thermostable group II intron reverse transcriptase fusion proteins and their use in cDNA synthesis and next-generation RNA sequencing. RNA 19, 958–970 (2013).

    Article  CAS  Google Scholar 

  17. Stamos, J. L., Lentzsch, A. M. & Lambowitz, A. M. Structure of a thermostable group II intron reverse transcriptase with template-primer and its functional and evolutionary implications. Mol. Cell 68, 926–939 (2017).

    Article  CAS  Google Scholar 

  18. Kim, Y. et al. Adenine base editing and prime editing of chemically derived hepatic progenitors rescue genetic liver disease. Cell Stem Cell 28, 1614–1624 (2021).

    Article  CAS  Google Scholar 

  19. Azuma, H. et al. Robust expansion of human hepatocytes in Fah–/–/Rag2–/–/Il2rg–/– mice. Nat. Biotechnol. 25, 903–910 (2007).

    Article  CAS  Google Scholar 

  20. Paulk, N. K. et al. Adeno-associated virus gene repair corrects a mouse model of hereditary tyrosinemia in vivo. Hepatology 51, 1200–1208 (2010).

    Article  CAS  Google Scholar 

  21. 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  Google Scholar 

  22. Roth, A. et al. A widespread self-cleaving ribozyme class is revealed by bioinformatics. Nat. Chem. Biol. 10, 56–60 (2014).

    Article  CAS  Google Scholar 

  23. Jin, S. et al. Genome-wide specificity of prime editors in plants. Nat. Biotechnol. 39, 1292–1299 (2021).

    Article  CAS  Google Scholar 

  24. Zhang, L. et al. Reverse-transcribed SARS-CoV-2 RNA can integrate into the genome of cultured human cells and can be expressed in patient-derived tissues. Proc. Natl Acad. Sci. USA 118, e2105968118 (2021).

  25. Peabody, D. S. The RNA binding site of bacteriophage MS2 coat protein. EMBO J. 12, 595–600 (1993).

    Article  CAS  Google Scholar 

  26. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  28. Kluesner, M. G. et al. EditR: a method to quantify base editing from Sanger sequencing. CRISPR J 1, 239–250 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  30. Xue, W. et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 (2007).

    Article  CAS  Google Scholar 

  31. Xue, W. et al. CRISPR-mediated direct mutation of cancer genes in the mouse liver. Nature 514, 380–385 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Wolfe, P. Zamore and members of the Xue and Sontheimer labs for helpful discussions. We thank Y. Liu in the University of Massachusetts Chan Medical School Morphology Core and G. Gao, Q. Su and J. Xie in the University of Massachusetts Chan Medical School Viral Vector Core for support. W.X. was supported by grants from the National Institutes of Health (DP2HL137167, P01HL131471, P01HL158506 and UG3HL147367), American Cancer Society (129056-RSG-16-093) 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., S.-Q.L. and Z.C. conducted molecular biological experiments. B.L. performed mouse work. B.L., X.D., H.C. and T.C.R. conducted high-throughput sequencing and bioinformatic analyses. B.L., X.D., W.X. and E.J.S. interpreted the data and wrote 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 a patent application on this work. The authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Jia Chen 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 MS2-PE2 and SunTag-PE2 design.

a, Schematic overview of MS2-PE2. The MS2 coat protein (MCP) was fused to the N terminus of M-MLV reverse transcriptase to enable recruitment by the MS2-pegRNAs. b, Sizes of Cas9 nickase and MCP-RT ORFs. c, Engineered MS2-pegRNAs with MS2 sequences appended into distinct sgRNA stem-loops, or onto the 3′ terminus. d, Schematic overview of SunTag-PE2. e, Schematics of scFv-RT and GCN4-Cas9 nickase. The scFv was fused to the N terminus of M-MLV RT (top). The 10xGCN4 epitope was fused to either the N terminus (SunTag-PE) or the C terminus of SpyCas9H840A (PE-SunTag).

Extended Data Fig. 2 Split-PE, SunTag-PE3 and MS2-PE3 tested in an mCherry reporter line and an endogenous locus.

a, A diagram of the mCherry reporter line that functions by converting a premature stop codon. b, Sequences of RTT and PBS, non-cognate (PBS + RTT), non-cognate PBS, and non-cognate RTT for the mCherry reporter line. c, Multiple MS2-pegRNAs tested in mCherry reporter cell lines. The pegRNA with MS2 on the repeat/anti-repeat stem-loop (pegRNA-1.1) has the highest editing efficiency (higher even than that of the original PE3) in this mCherry reporter line (n = 2). Therefore, the pegRNA1.1-Cas9H840A-MCP-RT system was designated as MS2-PE3. d, SunTag-PE3 and PE3-SunTag were tested in the mCherry reporter cell line. Two-tailed unpaired Student’s t-test: *P < 0.05 (n = 3). e, Sanger sequencing and EditR quantification of PE3, Split PE, SunTag-PE3 and MS2-PE3 by installing “CTT” at HEK3 sites in HEK293T cells. All plasmids were transfected at the same molar ratio. Genomic DNAs were isolated 72 h post transfection. f, Dose dependence of the RT-encoding plasmid. One microgram of H840A plasmid was co-transfected with plasmids encoding additional sPE components [pegRNA (0.3 µg), nicking sgRNA (0.1 µg), and RT (0.01-2 µg)] per well in a 12-well plate (n = 2). Data and error bars indicate the mean and standard deviation of two or three independent biological replicates, as indicated.

Extended Data Fig. 3 SunTag-PE3 and MS2-PE3 tested in reporter lines.

a, A diagram of the GFP reporter line that is activated by precise insertion of 18 bp (in place of a 39-bp non-functional sequence). Indels (+1) can restore mCherry expression. b, A diagram of the GFP reporter line that is activated by deletion of 47 bp; indels (+1) can restore mCherry expression. c, MS2-PE3 was tested in the GFP reporter line shown in panel a (n = 3). d, SunTag-PE3 was tested in the GFP reporter line shown in a (n = 3). e, MS2-PE3 was tested in the GFP reporter line shown in b (n = 3). Data and error bars indicate the mean and standard deviation of three independent biological replicates.

Extended Data Fig. 4 Amplicon sequencing of MS2-PE3 and SunTag-PE3 at multiple endogenous sites.

a, MS2-PE3 for editing by 1-bp substitution at multiple endogenous loci, including HEK3, RNF2, VEGFA, and FANCF in HEK293T cells. b, SunTag-PE3 for RNF2 and VEGFR editing to generate a 1-bp substitution in HEK293T cells. Two-tailed unpaired Student’s t-test: *P < 0.05, **P < 0.01. Data and error bars indicate the mean and standard deviation of three independent biological replicates.

Extended Data Fig. 5 Prime editing by alternative reverse transcriptases.

a, Illustration of prime editors with alternative RTs. Human codon-optimized E.r. maturase RT and GsI-IIC RT were cloned into the original PE2 in place of the M-MLV RT. b, Prime editing by alternative RT orthologs at the VEGFA site by 3-nt substitutions (+2 G to C and +4-5 GG to CT). Two-tailed unpaired Student’s t-test: *P < 0.05, **P < 0.01, ***P < 0.001. Data and error bars indicate the mean and standard deviation of three independent biological replicates.

Extended Data Fig. 6 Prime editing using mutant PE2 and sPE components.

Representative Sanger sequencing traces from prime editing experiments using mutant PE2 and sPE components. HEK293T cells were transfected with indicated plasmids, along with others encoding pegRNA and nicking sgRNA. Prime editing introduces a 3-nt substitution at the FANCF locus (+2 C to T and +4-5 TG to AC). The experiment was repeated two times. Sanger sequencing traces were analyzed by EditR.

Extended Data Fig. 7 Split PE2 enables genome editing in adult mice.

a, Representative images of tumors in liver with PE3 or split PE. Control group was pegRNA-injected only. b, Amplicon sequencing from representative animals using genomic DNA isolated from tumors.

Extended Data Fig. 8 Prime editing by separate RNA modules.

a, Schematic of proof-of-concept experiment on delivering the RT template separately. The 3’ extension of the pegRNA (the RTT-PBS sequence) was removed from the 3’ of the tracrRNA scaffold and provided separately under the control of a U6 promoter. An sgRNA plasmid was co-transfected to carry out the nicking event in conjunction with the nCas9. b, Illustration of the circularization pathway to generate petRNAs. c, PE efficiency by modular RNA components at the FANCF locus introducing a 3-nt substitution (+2 C to T and +4-5 TG to AC). Plasmids expressing RNAs were co-transfected with Cas9H840A and the split RT, which lacks the MCP domain. Nicking sgRNAs were used for all prime editing. d, Validation of petRNA adaptability to an alternative nickase. The petRNA was designed to target a site at the FANCF locus where SpyCas9 and SauCas9 nickases share the same nick and thus a single petRNA guide/primer/template sequence. The petRNA and the MCP-RT were co-transfected with plasmids encoding SpyCas9H840A-sgRNA or SauCas9N580A. Nicking sgRNAs were used for all prime editing. Two-tailed unpaired Student’s t-test: *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Data and error bars indicate the mean and standard deviation of three independent biological replicates.

Extended Data Fig. 9 Comparison of PE, sPE, and petRNA off-target effects at known Cas9 off-target sites of FANCF and HEK4 using deep sequencing.

On-target edits are shown in red and off-target edits are shown in green. Data and error bars indicate the mean and standard deviation of three independent biological replicates.

Extended Data Fig. 10 In vitro transcribed mRNA and purified RT protein used in the nucleofections, and FACS gating strategy.

a, Denaturing agarose gel analysis of the mRNAs produced in-house. The coding sequences of nCas9, MMLV-RT or PE2 were flanked by a capped 5’ UTR and a 3’ UTR, followed by a 110-nt poly(A) tract. b, SDS-PAGE analysis of the purified MMLV-RT protein. C, FACS gating examples for reporter cells.

Source data

Supplementary information

Supplementary Information

Supplementary Note, Tables 1–4 and sequences.

Reporting Summary

Source data

Source Data Fig. 3

Unprocessed gel and blot images for Fig. 3e.

Source Data Extended Data Fig. 10

Unprocessed gel and blot images for Extended Data Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, B., Dong, X., Cheng, H. et al. A split prime editor with untethered reverse transcriptase and circular RNA template. Nat Biotechnol 40, 1388–1393 (2022). https://doi.org/10.1038/s41587-022-01255-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01255-9

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