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. 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 resulting from the cellular response to dsDNA breaks. 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, 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, and that use a Cas9 nickase targeting the non-edited strand, manipulate the cellular DNA repair response to favour 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.

[1]  J. Tainer,et al.  Crystal structure of human uracil-DNA glycosylase in complex with a protein inhibitor: Protein mimicry of DNA , 1995, Cell.

[2]  Reuben S Harris,et al.  RNA editing enzyme APOBEC1 and some of its homologs can act as DNA mutators. , 2002, Molecular cell.

[3]  Yunmei Ma,et al.  Mechanism and regulation of human non-homologous DNA end-joining , 2003, Nature Reviews Molecular Cell Biology.

[4]  Reuben S Harris,et al.  Comparison of the differential context-dependence of DNA deamination by APOBEC enzymes: correlation with mutation spectra in vivo. , 2004, Journal of molecular biology.

[5]  R. Heller,et al.  Replisome assembly and the direct restart of stalled replication forks , 2006, Nature Reviews Molecular Cell Biology.

[6]  G. Forno,et al.  The Missing ApoE Allele , 2007, Annals of human genetics.

[7]  S. Conticello The AID/APOBEC family of nucleic acid mutators , 2008, Genome Biology.

[8]  Willem P C Stemmer,et al.  A recombinant polypeptide extends the in vivo half-life of peptides and proteins in a tunable manner , 2009, Nature Biotechnology.

[9]  D. Holtzman,et al.  The Role of Apolipoprotein E in Alzheimer's Disease , 2009, Neuron.

[10]  C. Kunz,et al.  DNA Repair in Mammalian Cells , 2009, Cellular and Molecular Life Sciences.

[11]  P. Modrich,et al.  PCNA function in the activation and strand direction of MutLα endonuclease in mismatch repair , 2010, Proceedings of the National Academy of Sciences.

[12]  Albert J R Heck,et al.  Structural basis for CRISPR RNA-guided DNA recognition by Cascade , 2011, Nature Structural &Molecular Biology.

[13]  A. Børresen-Dale,et al.  The landscape of cancer genes and mutational processes in breast cancer , 2012, Nature.

[14]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[15]  David R. Liu,et al.  High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity , 2013, Nature Biotechnology.

[16]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[17]  David A. Scott,et al.  Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.

[18]  V. Verkhusha,et al.  Near-infrared fluorescent proteins for multicolor in vivo imaging , 2013, Nature Methods.

[19]  G. Church,et al.  CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering , 2013, Nature Biotechnology.

[20]  Martin J. Aryee,et al.  Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing , 2014, Nature Biotechnology.

[21]  Mazhar Adli,et al.  Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease , 2014, Nature Biotechnology.

[22]  C. Sala,et al.  Erratum: The RNA editing enzyme APOBEC1 induces somatic mutations and a compatible mutational signature is present in esophageal adenocarcinomas , 2014, Genome Biology.

[23]  Jeffry D. Sander,et al.  CRISPR-Cas systems for editing, regulating and targeting genomes , 2014, Nature Biotechnology.

[24]  Randall J. Platt,et al.  Therapeutic genome editing: prospects and challenges , 2015, Nature Medicine.

[25]  Charles A Gersbach,et al.  Enabling functional genomics with genome engineering , 2015, Genome research.

[26]  David R. Liu,et al.  Small Molecule-Triggered Cas9 Protein with Improved Genome-Editing Specificity , 2015, Nature chemical biology.

[27]  Feng Zhang,et al.  Genome engineering using CRISPR-Cas9 system. , 2015, Methods in molecular biology.

[28]  Martin J. Aryee,et al.  GUIDE-Seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases , 2014, Nature Biotechnology.

[29]  Martin J. Aryee,et al.  Engineered CRISPR-Cas9 nucleases with altered PAM specificities , 2015, Nature.

[30]  J. Keith Joung,et al.  Efficient Delivery of Genome-Editing Proteins In Vitro and In Vivo , 2014, Nature Biotechnology.

[31]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[32]  Jennifer A. Doudna,et al.  Structures of a CRISPR-Cas9 R-loop complex primed for DNA cleavage , 2016, Science.

[33]  J. Keith Joung,et al.  731. High-Fidelity CRISPR-Cas9 Nucleases with No Detectable Genome-Wide Off-Target Effects , 2016 .

[34]  Ricardo Villamarín-Salomón,et al.  ClinVar: public archive of interpretations of clinically relevant variants , 2015, Nucleic Acids Res..