Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis

Recently developed DNA base editing methods enable the direct generation of desired point mutations in genomic DNA without generating any double-strand breaks1–3, but the issue of off-target edits has limited the application of these methods. Although several previous studies have evaluated off-target mutations in genomic DNA4–8, it is now clear that the deaminases that are integral to commonly used DNA base editors often bind to RNA9–13. For example, the cytosine deaminase APOBEC1—which is used in cytosine base editors (CBEs)—targets both DNA and RNA12, and the adenine deaminase TadA—which is used in adenine base editors (ABEs)—induces site-specific inosine formation on RNA9,11. However, any potential RNA mutations caused by DNA base editors have not been evaluated. Adeno-associated viruses are the most common delivery system for gene therapies that involve DNA editing; these viruses can sustain long-term gene expression in vivo, so the extent of potential RNA mutations induced by DNA base editors is of great concern14–16. Here we quantitatively evaluated RNA single nucleotide variations (SNVs) that were induced by CBEs or ABEs. Both the cytosine base editor BE3 and the adenine base editor ABE7.10 generated tens of thousands of off-target RNA SNVs. Subsequently, by engineering deaminases, we found that three CBE variants and one ABE variant showed a reduction in off-target RNA SNVs to the baseline while maintaining efficient DNA on-target activity. This study reveals a previously overlooked aspect of off-target effects in DNA editing and also demonstrates that such effects can be eliminated by engineering deaminases. Cytosine and adenine base editors have undesired off-target effects on RNA, but this activity can be reduced in deaminase-engineered variants while preserving on-target DNA editing.

[1]  Daesik Kim,et al.  Genome-wide target specificities of CRISPR RNA-guided programmable deaminases , 2017, Nature Biotechnology.

[2]  Haiyan Jiang,et al.  Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10 , 2019, Nature Medicine.

[3]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[4]  Li Li,et al.  In utero CRISPR-mediated therapeutic editing of metabolic genes , 2018, Nature Medicine.

[5]  Åsa K. Björklund,et al.  Full-length RNA-seq from single cells using Smart-seq2 , 2014, Nature Protocols.

[6]  Martin J. Aryee,et al.  Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors , 2019, Nature.

[7]  Rommie E. Amaro,et al.  Structural basis for targeted DNA cytosine deamination and mutagenesis by APOBEC3A and APOBEC3B , 2016, Nature Structural &Molecular Biology.

[8]  L. Poulsen,et al.  Analysis of an Escherichia coli mutant strain resistant to the cell‐killing function encoded by the gef gene family , 1992, Molecular microbiology.

[9]  Daesik Kim,et al.  Genome-wide target specificity of CRISPR RNA-guided adenine base editors , 2019, Nature Biotechnology.

[10]  Li Yang,et al.  Efficient base editing in methylated regions with a human APOBEC3A-Cas9 fusion , 2018, Nature Biotechnology.

[11]  Lukas Villiger,et al.  Treatment of a metabolic liver disease by in vivo genome base editing in adult mice , 2018, Nature Medicine.

[12]  H. Matsuo,et al.  Structure of the DNA deaminase domain of the HIV-1 restriction factor APOBEC3G , 2008, Nature.

[13]  M. Sowden,et al.  Overexpression of APOBEC-1 Results in Mooring Sequence-dependent Promiscuous RNA Editing (*) , 1996, The Journal of Biological Chemistry.

[14]  D. Graur,et al.  Inferring the Pattern of Spontaneous Mutation from the Pattern of Substitution in Unitary Pseudogenes of Mycobacterium leprae and a Comparison of Mutation Patterns Among Distantly Related Organisms , 2005, Journal of Molecular Evolution.

[15]  Q. Gao,et al.  Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice , 2019, Science.

[16]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

[17]  W. Keller,et al.  tadA, an essential tRNA‐specific adenosine deaminase from Escherichia coli , 2002, The EMBO journal.

[18]  David R. Liu,et al.  Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction , 2018, Nature Biotechnology.

[19]  David R. Liu,et al.  Analysis and minimization of cellular RNA editing by DNA adenine base editors , 2019, Science Advances.

[20]  G. Schneider,et al.  Model structure of APOBEC3C reveals a binding pocket modulating ribonucleic acid interaction required for encapsidation , 2009, Proceedings of the National Academy of Sciences.

[21]  Luca Pinello,et al.  An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities , 2018, Nature Biotechnology.

[22]  Fan Guo,et al.  Integrative single-cell analysis of transcriptome, DNA methylome and chromatin accessibility in mouse oocytes , 2018, Cell Research.

[23]  David R. Liu,et al.  Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage , 2016, Nature.

[24]  P. Green,et al.  Transcription-associated mutational asymmetry in mammalian evolution , 2003, Nature Genetics.

[25]  Kevin T. Zhao,et al.  Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions , 2017, Nature Biotechnology.

[26]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[27]  Emma M Schatoff,et al.  Optimized base editors enable efficient editing in cells, organoids and mice , 2018, Nature Biotechnology.

[28]  R. Wolfenden,et al.  The structure of the cytidine deaminase-product complex provides evidence for efficient proton transfer and ground-state destabilization. , 1997, Biochemistry.

[29]  V. Schramm,et al.  Structural and kinetic characterization of Escherichia coli TadA, the wobble-specific tRNA deaminase. , 2006, Biochemistry.

[30]  L. Steinmetz,et al.  Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos , 2019, Science.

[31]  Martin J. Aryee,et al.  CRISPR adenine and cytosine base editors with reduced RNA off-target activities , 2019, bioRxiv.

[32]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[33]  David R. Liu,et al.  Base editing: precision chemistry on the genome and transcriptome of living cells , 2018, Nature Reviews Genetics.

[34]  G. Crooks,et al.  WebLogo: a sequence logo generator. , 2004, Genome research.

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

[36]  R. Stevens,et al.  Crystal structure of the anti-viral APOBEC3G catalytic domain and functional implications , 2008, Nature.

[37]  N. Davidson,et al.  APOBEC‐1‐mediated RNA editing , 2010, Wiley interdisciplinary reviews. Systems biology and medicine.

[38]  M. DePristo,et al.  The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. , 2010, Genome research.

[39]  L. Duret Mutation Patterns in the Human Genome: More Variable Than Expected , 2009, PLoS biology.