16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Efficient CRISPR editing with a hypercompact Cas12f1 and engineered guide RNAs delivered by adeno-associated virus

      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

          Gene therapy would benefit from a miniature CRISPR system that fits into the small adeno-associated virus (AAV) genome and has high cleavage activity and specificity in eukaryotic cells. One of the most compact CRISPR-associated nucleases yet discovered is the archaeal Un1Cas12f1. However, Un1Cas12f1 and its variants have very low activity in eukaryotic cells. In the present study, we redesigned the natural guide RNA of Un1Cas12f1 at five sites: the 5′ terminus of the trans-activating CRISPR RNA (tracrRNA), the tracrRNA–crRNA complementary region, a penta(uridinylate) sequence, the 3′ terminus of the crRNA and a disordered stem 2 region in the tracrRNA. These optimizations synergistically increased the average indel frequency by 867-fold. The optimized Un1Cas12f1 system enabled efficient, specific genome editing in human cells when delivered by plasmid vectors, PCR amplicons and AAV. As Un1Cas12f1 cleaves outside the protospacer, it can be used to create large deletions efficiently. The engineered Un1Cas12f1 system showed efficiency comparable to that of SpCas9 and specificity similar to that of AsCas12a.

          Abstract

          A miniature CRISPR system may enable genome editing using single AAV vectors.

          Related collections

          Most cited references51

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

          A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.

          Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems provide bacteria and archaea with adaptive immunity against viruses and plasmids by using CRISPR RNAs (crRNAs) to guide the silencing of invading nucleic acids. We show here that in a subset of these systems, the mature crRNA that is base-paired to trans-activating crRNA (tracrRNA) forms a two-RNA structure that directs the CRISPR-associated protein Cas9 to introduce double-stranded (ds) breaks in target DNA. At sites complementary to the crRNA-guide sequence, the Cas9 HNH nuclease domain cleaves the complementary strand, whereas the Cas9 RuvC-like domain cleaves the noncomplementary strand. The dual-tracrRNA:crRNA, when engineered as a single RNA chimera, also directs sequence-specific Cas9 dsDNA cleavage. Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA-programmable genome editing.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage

            Current genome-editing technologies introduce double-stranded (ds) DNA breaks at a target locus as the first step to gene correction. 1,2 Although most genetic diseases arise from point mutations, current approaches to point mutation correction are inefficient and typically induce an abundance of random insertions and deletions (indels) at the target locus from the cellular response to dsDNA breaks. 1,2 Here we report the development of base editing, a new approach to genome editing that enables the direct, irreversible conversion of one target DNA base into another in a programmable manner, without requiring dsDNA backbone cleavage or a donor template. We engineered fusions of CRISPR/Cas9 and a cytidine deaminase enzyme that retain the ability to be programmed with a guide RNA, do not induce dsDNA breaks, and mediate the direct conversion of cytidine to uridine, thereby effecting a C→T (or G→A) substitution. The resulting “base editors” convert cytidines within a window of approximately five nucleotides (nt), and can efficiently correct a variety of point mutations relevant to human disease. In four transformed human and murine cell lines, second- and third-generation base editors that fuse uracil glycosylase inhibitor (UGI), and that use a Cas9 nickase targeting the non-edited strand, manipulate the cellular DNA repair response to favor desired base-editing outcomes, resulting in permanent correction of ∼15-75% of total cellular DNA with minimal (typically ≤ 1%) indel formation. Base editing expands the scope and efficiency of genome editing of point mutations.
              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

                Author and article information

                Contributors
                omsys1@kribb.re.kr
                Journal
                Nat Biotechnol
                Nat Biotechnol
                Nature Biotechnology
                Nature Publishing Group US (New York )
                1087-0156
                1546-1696
                2 September 2021
                2 September 2021
                2022
                : 40
                : 1
                : 94-102
                Affiliations
                [1 ]GRID grid.249967.7, ISNI 0000 0004 0636 3099, Genome Editing Research Center, , Korea Research Institute of Bioscience & BioTechnology, ; Daejeon, Republic of Korea
                [2 ]GRID grid.412786.e, ISNI 0000 0004 1791 8264, KRIBB School of Bioscience, , Korea University of Science and Technology, ; Daejeon, Republic of Korea
                [3 ]GenKOre, Daejeon, Republic of Korea
                [4 ]GRID grid.289247.2, ISNI 0000 0001 2171 7818, Department of Biomedical and Pharmaceutical Sciences, , Graduate School, Kyung Hee University, ; Seoul, Republic of Korea
                [5 ]GRID grid.289247.2, ISNI 0000 0001 2171 7818, Department of Pharmaceutical Science, College of Pharmacy, , Kyung Hee University, ; Seoul, Republic of Korea
                Author information
                http://orcid.org/0000-0001-6829-2802
                http://orcid.org/0000-0003-0806-5979
                http://orcid.org/0000-0002-3133-3954
                Article
                1009
                10.1038/s41587-021-01009-z
                8763643
                34475560
                5f8af8da-9071-4862-b306-e885dba68ac0
                © The Author(s) 2021

                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
                : 25 January 2021
                : 8 July 2021
                Funding
                Funded by: FundRef https://doi.org/10.13039/501100003052, Ministry of Trade, Industry and Energy (Ministry of Trade, Industry and Energy, Korea);
                Award ID: 20012445
                Award ID: 20012445
                Award ID: 20012445
                Award ID: 20012445
                Award ID: 20012445
                Award Recipient :
                Funded by: - Commercializations Promotion Agency For R&D Outcomes funded by the Ministry of Science and ICT (1711121245).
                Funded by: - KRIBB research Initiative Program funded by the Ministry of Science and ICT.
                Funded by: - KRIBB research Initiative Program funded by the Ministry of Science and ICT. - Commercializations Promotion Agency For R&D Outcomes funded by the Ministry of Science and ICT (1711121245).
                Categories
                Article
                Custom metadata
                © The Author(s), under exclusive licence to Springer Nature America, Inc. 2022

                Biotechnology
                targeted gene repair,crispr-cas9 genome editing
                Biotechnology
                targeted gene repair, crispr-cas9 genome editing

                Comments

                Comment on this article