18
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Prediction of prime editing insertion efficiencies using sequence features and DNA repair determinants

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Most short sequences can be precisely written into a selected genomic target using prime editing; however, it remains unclear what factors govern insertion. We design a library of 3,604 sequences of various lengths and measure the frequency of their insertion into four genomic sites in three human cell lines, using different prime editor systems in varying DNA repair contexts. We find that length, nucleotide composition and secondary structure of the insertion sequence all affect insertion rates. We also discover that the 3′ flap nucleases TREX1 and TREX2 suppress the insertion of longer sequences. Combining the sequence and repair features into a machine learning model, we can predict relative frequency of insertions into a site with R = 0.70. Finally, we demonstrate how our accurate prediction and user-friendly software help choose codon variants of common fusion tags that insert at high efficiency, and provide a catalog of empirically determined insertion rates for over a hundred useful sequences.

          Abstract

          Prime editing efficiency is affected by insertion sequence features, secondary structure and DNA repair context.

          Related collections

          Most cited references60

          • Record: found
          • Abstract: not found
          • Conference Proceedings: not found

          XGBoost

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Search-and-replace genome editing without double-strand breaks or donor DNA

            Summary Most genetic variants that contribute to disease 1 are challenging to correct efficiently and without excess byproducts 2–5 . Here we describe prime editing, a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a catalytically impaired Cas9 fused to an engineered reverse transcriptase, programmed with a prime editing guide RNA (pegRNA) that both specifies the target site and encodes the desired edit. We performed >175 edits in human cells including targeted insertions, deletions, and all 12 types of point mutations without requiring double-strand breaks or donor DNA templates. We applied prime editing in human cells to correct efficiently and with few byproducts the primary genetic causes of sickle cell disease (requiring a transversion in HBB) and Tay-Sachs disease (requiring a deletion in HEXA), to install a protective transversion in PRNP, and to precisely insert various tags and epitopes into target loci. Four human cell lines and primary post-mitotic mouse cortical neurons support prime editing with varying efficiencies. Prime editing shows higher or similar efficiency and fewer byproducts than homology-directed repair, complementary strengths and weaknesses compared to base editing, and much lower off-target editing than Cas9 nuclease at known Cas9 off-target sites. Prime editing substantially expands the scope and capabilities of genome editing, and in principle can correct up to 89% of known genetic variants associated with human diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9

              CRISPR-Cas9-based genetic screens are a powerful new tool in biology. By simply altering the sequence of the single-guide RNA (sgRNA), Cas9 can be reprogrammed to target different sites in the genome with relative ease, but the on-target activity and off-target effects of individual sgRNAs can vary widely. Here, we use recently-devised sgRNA design rules to create human and mouse genome-wide libraries, perform positive and negative selection screens and observe that the use of these rules produced improved results. Additionally, we profile the off-target activity of thousands of sgRNAs and develop a metric to predict off-target sites. We incorporate these findings from large-scale, empirical data to improve our computational design rules and create optimized sgRNA libraries that maximize on-target activity and minimize off-target effects to enable more effective and efficient genetic screens and genome engineering.
                Bookmark

                Author and article information

                Contributors
                leopold.parts@sanger.ac.uk
                Journal
                Nat Biotechnol
                Nat Biotechnol
                Nature Biotechnology
                Nature Publishing Group US (New York )
                1087-0156
                1546-1696
                16 February 2023
                16 February 2023
                2023
                : 41
                : 10
                : 1446-1456
                Affiliations
                [1 ]Wellcome Sanger Institute, ( https://ror.org/05cy4wa09) Hinxton, UK
                [2 ]Department of Computer Science, University of Tartu, ( https://ror.org/03z77qz90) Tartu, Estonia
                Author information
                http://orcid.org/0000-0003-1306-3994
                http://orcid.org/0000-0002-1310-6168
                http://orcid.org/0000-0002-4840-195X
                http://orcid.org/0000-0002-0192-5385
                http://orcid.org/0000-0002-2618-670X
                Article
                1678
                10.1038/s41587-023-01678-y
                10567557
                36797492
                21928715-db8e-4e28-8322-802aaab78790
                © The Author(s) 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 10 November 2021
                : 18 January 2023
                Funding
                Funded by: FundRef https://doi.org/10.13039/100004440, Wellcome Trust (Wellcome);
                Award ID: 108413/A/15/D
                Award ID: 220540/Z/20/A
                Award ID: 108413/A/15/D
                Award ID: 108413/A/15/D
                Award ID: 108413/A/15/D
                Award ID: 108413/A/15/D
                Award ID: 220540/Z/20/A
                Award ID: 108413/A/15/D
                Award ID: 108413/A/15/D
                Award ID: 220540/Z/20/A
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100008530, EC | European Regional Development Fund (Europski Fond za Regionalni Razvoj);
                Award ID: 2014-2020.4.01.16-0271
                Award ID: 2014-2020.4.01.16-0271
                Award ID: 2014-2020.4.01.16-0271
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100002301, Eesti Teadusagentuur (Estonian Research Council);
                Award ID: TT11
                Award ID: TT11
                Award ID: TT11
                Award Recipient :
                Funded by: Estonian Centre of Excellence in IT (EXCITE) (TK148)
                Categories
                Article
                Custom metadata
                © Springer Nature America, Inc. 2023

                Biotechnology
                functional genomics,synthetic biology,computational models,genetic engineering,high-throughput screening

                Comments

                Comment on this article