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.

Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors

Abstract

CRISPR–Cas base-editor technology enables targeted nucleotide alterations, and is being increasingly used for research and potential therapeutic applications1,2. The most widely used cytosine base editors (CBEs) induce deamination of DNA cytosines using the rat APOBEC1 enzyme, which is targeted by a linked Cas protein–guide RNA complex3,4. Previous studies of the specificity of CBEs have identified off-target DNA edits in mammalian cells5,6. Here we show that a CBE with rat APOBEC1 can cause extensive transcriptome-wide deamination of RNA cytosines in human cells, inducing tens of thousands of C-to-U edits with frequencies ranging from 0.07% to 100% in 38–58% of expressed genes. CBE-induced RNA edits occur in both protein-coding and non-protein-coding sequences and generate missense, nonsense, splice site, and 5′ and 3′ untranslated region mutations. We engineered two CBE variants bearing mutations in rat APOBEC1 that substantially decreased the number of RNA edits (by more than 390-fold and more than 3,800-fold) in human cells. These variants also showed more precise on-target DNA editing than the wild-type CBE and, for most guide RNAs tested, no substantial reduction in editing efficiency. Finally, we show that an adenine base editor7 can also induce transcriptome-wide RNA edits. These results have implications for the use of base editors in both research and clinical settings, illustrate the feasibility of engineering improved variants with reduced RNA editing activities, and suggest the need to more fully define and characterize the RNA off-target effects of deaminase enzymes in base editor platforms.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Transcriptome-wide off-target C-to-U RNA editing induced by BE3 in HepG2 cells.
Fig. 2: SECURE BE3 variants with substantially reduced RNA editing activities but comparable and more-precise DNA editing in HEK293T cells.
Fig. 3: ABEmax induces transcriptome-wide off-target A-to-I RNA editing in HEK293T cells.

Similar content being viewed by others

Data availability

Plasmids encoding the most relevant constructs shown in this work, including both SECURE BE3 variants, have been deposited to Addgene (http://www.addgene.org/browse/article/28197497/; Addgene IDs 123611–123616).

All RNA-seq data used in this study have been deposited in the Gene Expression Omnibus (GEO) repository (National Center for Biotechnology Information). The files are accessible through the GEO Series accession number GSE121668. All WES and targeted amplicon sequencing data have been deposited at the SRA repository under bioproject number PRJNA497753. All other relevant data are available from the corresponding author on request.

Code availability

The authors will make all previously unreported custom computer code used in this work available upon reasonable request.

References

  1. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770–788 (2018).

    Article  CAS  Google Scholar 

  2. Seo, H. & Kim, J. S. Towards therapeutic base editing. Nat. Med. 24, 1493–1495 (2018).

    Article  Google Scholar 

  3. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).

    Article  ADS  CAS  Google Scholar 

  4. Komor, A. C. et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci. Adv. 3, eaao4774 (2017).

    Google Scholar 

  5. Kim, D. et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat. Biotechnol. 35, 475–480 (2017).

    Article  CAS  Google Scholar 

  6. Zuo, E. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289–292 (2019).

    Chapter  Google Scholar 

  7. Gaudelli, N. M. et al. Programmable base editing of A·T to G·C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  ADS  CAS  Google Scholar 

  8. Salter, J. D., Bennett, R. P. & Smith, H. C. The APOBEC protein family: united by structure, divergent in function. Trends Biochem. Sci. 41, 578–594 (2016).

    Article  CAS  Google Scholar 

  9. Harris, R. S., Petersen-Mahrt, S. K. & Neuberger, M. S. RNA editing enzyme APOBEC1 and some of its homologs can act as DNA mutators. Mol. Cell 10, 1247–1253 (2002).

    Article  CAS  Google Scholar 

  10. Lau, P. P., Chen, S. H., Wang, J. C. & Chan, L. A 40 kilodalton rat liver nuclear protein binds specifically to apolipoprotein B mRNA around the RNA editing site. Nucleic Acids Res. 18, 5817–5821 (1990).

    Article  CAS  Google Scholar 

  11. Boström, K. et al. Apolipoprotein B mRNA editing. Direct determination of the edited base and occurrence in non-apolipoprotein B-producing cell lines. J. Biol. Chem. 265, 22446–22452 (1990).

    PubMed  Google Scholar 

  12. Skuse, G. R., Cappione, A. J., Sowden, M., Metheny, L. J. & Smith, H. C. The neurofibromatosis type I messenger RNA undergoes base-modification RNA editing. Nucleic Acids Res. 24, 478–485 (1996).

    Article  CAS  Google Scholar 

  13. Rosenberg, B. R., Hamilton, C. E., Mwangi, M. M., Dewell, S. & Papavasiliou, F. N. Transcriptome-wide sequencing reveals numerous APOBEC1 mRNA-editing targets in transcript 3′ UTRs. Nat. Struct. Mol. Biol. 18, 230–236 (2011).

    Article  CAS  Google Scholar 

  14. Sowden, M., Hamm, J. K. & Smith, H. C. Overexpression of APOBEC-1 results in mooring sequence-dependent promiscuous RNA editing. J. Biol. Chem. 271, 3011–3017 (1996).

    Article  CAS  Google Scholar 

  15. Yamanaka, S., Poksay, K. S., Driscoll, D. M. & Innerarity, T. L. Hyperediting of multiple cytidines of apolipoprotein B mRNA by APOBEC-1 requires auxiliary protein(s) but not a mooring sequence motif. J. Biol. Chem. 271, 11506–11510 (1996).

    Article  CAS  Google Scholar 

  16. Powell, L. M. et al. A novel form of tissue-specific RNA processing produces apolipoprotein-B48 in intestine. Cell 50, 831–840 (1987).

    Article  CAS  Google Scholar 

  17. Chen, S. H. et al. Apolipoprotein B-48 is the product of a messenger RNA with an organ-specific in-frame stop codon. Science 238, 363–366 (1987).

    Article  ADS  CAS  Google Scholar 

  18. Yamanaka, S., Poksay, K. S., Balestra, M. E., Zeng, G. Q. & Innerarity, T. L. Cloning and mutagenesis of the rabbit ApoB mRNA editing protein. A zinc motif is essential for catalytic activity, and noncatalytic auxiliary factor(s) of the editing complex are widely distributed. J. Biol. Chem. 269, 21725–21734 (1994).

    CAS  PubMed  Google Scholar 

  19. Navaratnam, N. et al. Evolutionary origins of apoB mRNA editing: catalysis by a cytidine deaminase that has acquired a novel RNA-binding motif at its active site. Cell 81, 187–195 (1995).

    Article  CAS  Google Scholar 

  20. Teng, B. B. et al. Mutational analysis of apolipoprotein B mRNA editing enzyme (APOBEC1). Structure–function relationships of RNA editing and dimerization. J. Lipid Res. 40, 623–635 (1999).

    CAS  PubMed  Google Scholar 

  21. Chen, Z. et al. Hypermutation induced by APOBEC-1 overexpression can be eliminated. RNA 16, 1040–1052 (2010).

    Article  CAS  Google Scholar 

  22. MacGinnitie, A. J., Anant, S. & Davidson, N. O. Mutagenesis of apobec-1, the catalytic subunit of the mammalian apolipoprotein B mRNA editing enzyme, reveals distinct domains that mediate cytosine nucleoside deaminase, RNA binding, and RNA editing activity. J. Biol. Chem. 270, 14768–14775 (1995).

    Article  CAS  Google Scholar 

  23. Wolf, J., Gerber, A. P. & Keller, W. tadA, an essential tRNA-specific adenosine deaminase from Escherichia coli. EMBO J. 21, 3841–3851 (2002).

    Article  CAS  Google Scholar 

  24. Kim, J. et al. Structural and kinetic characterization of Escherichia coli TadA, the wobble-specific tRNA deaminase. Biochemistry 45, 6407–6416 (2006).

    Article  CAS  Google Scholar 

  25. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36, 843–846 (2018).

    Article  CAS  Google Scholar 

  26. Gibson, D. G. et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 (2009).

    Article  CAS  Google Scholar 

  27. Laird, P. W. et al. Simplified mammalian DNA isolation procedure. Nucleic Acids Res. 19, 4293 (1991).

    Article  CAS  Google Scholar 

  28. Rohland, N. & Reich, D. Cost-effective, high-throughput DNA sequencing libraries for multiplexed target capture. Genome Res. 22, 939–946 (2012).

    Article  CAS  Google Scholar 

  29. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  30. DePristo, M. A. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  Google Scholar 

  31. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  Google Scholar 

  32. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  Google Scholar 

  33. Olarerin-George, A. O. & Hogenesch, J. B. Assessing the prevalence of mycoplasma contamination in cell culture via a survey of NCBI’s RNA-seq archive. Nucleic Acids Res. 43, 2535–2542 (2015).

    Article  CAS  Google Scholar 

  34. McLaren, W. et al. The Ensembl variant effect predictor. Genome Biol. 17, 122 (2016).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  36. Crooks, G. E., Hon, G., Chandonia, J. M. & Brenner, S. E. WebLogo: a sequence logo generator. Genome Res. 14, 1188–1190 (2004).

    Article  CAS  Google Scholar 

  37. Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N. & Sternberg, M. J. The Phyre2 web portal for protein modeling, prediction and analysis. Nat. Protocols 10, 845–858 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

J.K.J., J.G. and R.Z. are supported by the Defense Advanced Research Projects Agency (HR0011-17-2-0042). Support was also provided by the National Institutes of Health (RM1 HG009490 to J.K.J. and J.G. and R35 GM118158 to J.K.J. and M.J.A.). J.K.J. is additionally supported by the Desmond and Ann Heathwood MGH Research Scholar Award. We thank M. M. Kaminski, B. P. Kleinstiver and K. Petri for discussions; V. Pattanayak for input on the manuscript; Y. E. Tak, G. Boulay, M. K. Clement, A. A. Sousa, R. T. Walton, M. L. Bobbin, M. V. Maus and A. Schmidts for technical advice; and P. K. Cabeceiras and O. R. Cervantes for technical assistance. J.K.J. dedicates this paper to the memory of C. J. Park.

Author information

Authors and Affiliations

Authors

Contributions

J.G. and R.Z. performed all wet laboratory experiments together. S.P.G., S.I., C.A.L. and M.J.A. performed all bioinformatic and computational analysis of data. J.G. and J.K.J. conceived and designed the study. J.G., M.J.A. and J.K.J. organized and supervised the work. J.G. and J.K.J. wrote the initial draft of the manuscript and all authors contributed to the writing of the final manuscript.

Corresponding author

Correspondence to J. Keith Joung.

Ethics declarations

Competing interests

J.K.J. has financial interests in Beam Therapeutics, Editas Medicine, Endcadia, Pairwise Plants, Poseida Therapeutics and Transposagen Biopharmaceuticals. These interests were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. J.K.J. and M.J.A. also hold equity in Excelsior Genomics. J.K.J. is a member of the Board of Directors of the American Society of Gene and Cell Therapy. J.G., R.Z. and J.K.J. are co-inventors on patent applications that have been filed by Partners Healthcare/Massachusetts General Hospital on engineered base editor architectures that reduce RNA editing activities.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Additional data and analysis for transcriptome-wide off-target C-to-U RNA editing induced with BE3 in HepG2 cells.

a, Dot plot of RNF2 on-target DNA editing data shown in Fig. 1b, depicting editing frequencies for all cytosines across the spacer sequence. b, Heat maps showing RNA and DNA editing efficiencies with BE3 and control on cytosines in human APOB. Numbering indicates nucleotide positions in the APOB transcript; asterisks identify those previously shown to be modified by APOBEC1. c, Histograms showing numbers of RNA-edited cytosines identified (y-axis) with RNA C-to-U editing frequencies (x-axis) for the four replicates shown in Fig. 1c. Dashed red line, median; solid red line, mean. d, Manhattan plots of data for replicates 2, 3, and 4 from Fig. 1c showing the distribution of modified cytosines identified across the transcriptome. n, total number of modified cytosines identified. e, Percentages of different predicted effects and locations of edited cytosines identified in each RNA-seq replicate. f, Jitter plots of cytosines modified by BE3 expression with the RNF2 gRNA categorized by their presence in 4, 3, 2 or 1 of the replicate RNA-seq experiments performed in HepG2 cells (n = 4 biologically independent samples, as in Fig. 1c). Box spans the interquartile range (IQR) (first to third quartiles); horizontal line shows median (second quartile); whiskers extend to ± 1.5 × IQR. n, total number of modified cytosines present in each category. The percentage of all modified cytosines in each category is also shown.

Extended Data Fig. 2 BE3 expression with two different gRNAs induces transcriptome-wide off-target RNA editing in HEK293T cells.

a, Heat maps of on-target DNA base editing efficiencies of BE3 and nCas9–UGI–NLS (control) in HEK293T cells (all GFP sorting) determined in triplicate with RNF2 or EMX1 gRNA. Bases shown are within the editing window of the on-target spacer sequence (numbering is at the bottom with 1 being the most PAM-distal spacer position). b, Dot plots of RNF2 and EMX1 on-target DNA editing data shown in a, depicting editing frequencies for all cytosines across the spacer sequence. c, Jitter plots derived from RNA-seq experiments showing RNA cytosines modified by BE3 expression with RNF2 or EMX1 gRNA. n, total number of modified cytosines identified in each replicate. d, Histograms showing numbers of RNA-edited cytosines identified (y-axis) with RNA C-to-U editing frequencies (x-axis) for the experiments shown in c. Dashed red line, median; solid red line, mean. e, Manhattan plots of data shown in c depicting the distribution of modified cytosines across the transcriptome. n, total number of modified cytosines identified.

Extended Data Fig. 3 Additional analysis of data showing transcriptome-wide off-target RNA editing in HEK293T cells with BE3 and two different gRNAs.

a, Percentages of different predicted effects and locations of edited cytosines in each RNA-seq replicate from Extended Data Fig. 2c. b, Percentages (x-axis) and numbers (shown inside bars) of expressed genes in each RNA-seq replicate from data shown in Extended Data Fig. 2c that show at least one edited cytosine. c, Jitter plots of cytosines modified by BE3 expression with RNF2 or EMX1 gRNA categorized by their presence in 3, 2 or 1 of the replicate RNA-seq experiments performed in HEK293T cells (n = 3 biologically independent samples, as in Extended Data Fig. 2c). Box, whiskers and n are as defined in Extended Data Fig. 1f. The percentage of all modified cytosines identified in each category is also shown. d, Sequence logos derived from edited cytosines identified in each RNA-seq replicate. Analysis done using RNA-seq data generated from cDNA; every T depicted should be considered a U in RNA. e, Venn diagram showing numbers of cytosines edited with the RNF2 and EMX1 gRNAs. For each gRNA, the number of cytosines represents the union of those identified in the three replicates.

Extended Data Fig. 4 Increased BE3 expression induces higher numbers and frequencies of transcriptome-wide RNA cytosine edits in HEK293T cells.

a, Heat maps of on-target DNA base editing efficiencies of BE3 and nCas9–UGI–NLS (control) in HEK293T cells (top 5% GFP sorting) determined in duplicate with RNF2 or EMX1 gRNA. Bases shown are within the editing window of the on-target spacer sequence (numbering is at the bottom with 1 being the most PAM-distal spacer position). b, Dot plots of data shown in a, depicting editing frequencies for all cytosines across the spacer sequence. c, Jitter plots derived from duplicate RNA-seq experiments showing RNA cytosines modified by BE3 expression with RNF2, EMX1 or non-targeted (NT) gRNA. n, total number of modified cytosines identified in each replicate. d, Histograms showing numbers of RNA edited cytosines identified (y-axis) with RNA C-to-U editing frequencies (x-axis) for the experiments shown in c. Dashed red line, median; solid red line, mean. e, Manhattan plots of data for both replicates of RNF2, EMX1, and non-targeted gRNAs from c showing the distribution of modified cytosines across the transcriptome. n,  total number of modified cytosines identified.

Extended Data Fig. 5 Additional data and analysis showing that increased BE3 expression induces higher numbers and frequencies of transcriptome-wide RNA cytosine edits in HEK293T cells.

a, Percentages of different predicted effects and locations of edited cytosines identified in each RNA-seq replicate from Extended Data Fig. 4c. b, Percentages (x-axis) and numbers (shown inside bars) of expressed genes in each RNA-seq replicate that have at least one edited cytosine. c, Sequence logos derived from edited cytosines identified in each RNA-seq duplicate experiment from Extended Data Fig. 4c for the RNF2, EMX1 and non-targeted gRNAs. Analysis done using RNA-seq data generated from cDNA; every T depicted should be considered a U in RNA. d, Venn diagram showing numbers of edited cytosines identified with the RNF2, EMX1 and non-targeted gRNAs. For each gRNA, the circle encompasses the union of cytosines identified in the two replicates (data derived from the experiments shown in Extended Data Fig. 4c). e, Venn diagrams showing all possible pairwise comparisons of edited cytosines identified in duplicate experiments performed with the RNF2, EMX1 and non-targeted gRNAs (data derived from the experiments shown in Extended Data Fig. 4c). f, Scatter plot correlating RNA editing frequencies (x-axis) of 154,264 cytosines previously shown to be edited by RNA-seq with DNA editing frequencies (y-axis) determined by WES performed with DNA derived from the same experiments (n = 3 biologically independent samples, pooled data). Superimposed histograms (top and right) depict the percentages of cytosines that show various editing rates on RNA (upper x-axis) or DNA (right y-axis).

Extended Data Fig. 6 Additional data showing that SECURE BE3 variants induce substantially reduced numbers of RNA edits but possess comparable and more-precise DNA editing activities in HEK293T cells.

a, Initial screen of transcriptome-wide RNA editing activities of six BE3 variants containing various rAPOBEC1 mutations and expressed at high levels in HEK293T cells (sorting cells with top 5% of GFP signal). Jitter plots of single replicate RNA-seq experiments showing RNA cytosines modified by expression of wild-type BE3, BE3-E63Q (rAPOBEC1 catalytic site mutant), BE3-P29F, BE3-P29T, BE3-L182A, BE3-R33A, BE3-K34A and BE3-R33A/K34A variants. n, total number of modified cytosines identified in each sample. b, Heat map of on-target DNA base-editing efficiencies of nCas9–UGI–NLS (control), wild-type BE3, BE3-R33A and BE3-R33A/K34A in HEK293T cells with the RNF2 gRNA (cells from experiment shown in Fig. 2a). Bases within the editing window of the on-target spacer sequence are numbered as previously described. Note the inclusion of C12, which is inefficiently edited by wild-type BE3 in these samples but not edited by the SECURE BE3 variants, even with higher expression. c, Dot plot for HEK293T on-target data displayed in b, expanded to include all cytosines across the spacer sequence.

Extended Data Fig. 7 Additional data and analysis of the on-target DNA and off-target RNA activities of BE3 and SECURE BE3 variants.

a, Dot plots illustrating on-target DNA editing efficiencies of nCas9–UGI–NLS (control), wild-type BE3, BE3-R33A and BE3-R33A/K34A in HEK293T cells on 12 genomic sites. These are the same data as shown in Fig. 2c, expanded to include all cytosines across the spacer sequence. b, Jitter plots from RNA-seq experiments in HepG2 cells showing RNA cytosines modified by wild-type BE3, BE3-R33A and BE3-R33A/K34A. Data for wild-type BE3 are from the experiments presented in Fig. 1c (replicates 2–4). n, total number of modified cytosines identified. c, Manhattan plots of data showing the distribution of modified cytosines induced with BE3-R33A or BE3-R33A/K34A expression for replicate 3 from b overlaid on modified cytosines induced with wild-type BE3 expression (the wild-type BE3 data are the same in the top and bottom plots). n, total number of modified cytosines identified. d, Heat map of on-target DNA base editing efficiencies of nCas9–UGI–NLS (control), wild-type BE3, BE3-R33A and BE3-R33A/K34A in HepG2 cells with the RNF2 gRNA (cells from same experiment as shown in b). Replicates 1, 2 and 3 for wild-type BE3 and nCas9–UGI–NLS show the same data presented as replicates 2, 3 and 4 for wild-type BE3 and nCas9–UGI–NLS in Fig. 1b. Bases within the editing window of the on-target spacer sequence are numbered as previously described. Note again the inclusion of position C12. e, Dot plot for HepG2 on-target data shown in d, expanded to include all cytosines across the spacer sequence. f, Schematic of the editing windows (coloured boxes) for wild-type BE3, BE3-R33A and BE3-R33A/K34A based on experimental data from Fig. 2c and Extended Data Fig. 7a. Darker-coloured and more-translucent boxes indicate positions generally showing higher and lower C-to-T editing efficiencies, respectively. Increased stringency for a 5′T with BE3-R33A/K34A is also indicated. The PAM (NGG) and the nicking site in the DNA backbone are highlighted. Drawings are adapted with permission from table 1 of ref. 1.

Extended Data Fig. 8 Additional data and analysis for transcriptome-wide off-target A-to-I RNA editing induced by ABEmax expression in HEK293T cells.

a, Dot plot of HEK site 2 on-target DNA editing data shown in Fig. 3a, depicting editing frequencies for all adenines across the spacer sequence. b, Histograms showing numbers of RNA-edited adenines identified (y-axis) with RNA A-to-I editing frequencies (x-axis) for three replicates shown in Fig. 3b. Dashed red line, median; solid red line, mean. c, Manhattan plots of data for replicates 1 and 2 from Fig. 3b showing the distribution of modified adenines identified across the transcriptome. n, total number of modified adenines identified. d, Percentages of different predicted effects and locations of edited adenines in each RNA-seq replicate shown in Fig. 3b. e, Percentages (x-axis) and numbers (inside bars) of expressed genes in each RNA-seq replicate that show at least one edited adenine. f, Jitter plots of adenines modified by ABEmax expression with the HEK site 2 gRNA categorized by their presence in 3, 2 or 1 of the replicate RNA-seq experiments shown in Fig. 3b (n = 3 biologically independent samples). Box and whiskers are as defined in Extended Data Fig. 1f. n, total number of modified adenines present in each category. The percentage of all modified adenines found in each category is also shown. g, Scatter plot correlating RNA editing frequencies (x-axis) of 52,462 adenines previously shown to be RNA edited with DNA editing frequencies (y-axis) determined by WES (n = 3 biologically independent samples, pooled data). Superimposed histograms (top and right) depict the percentages of edited adenines on RNA (upper x-axis) or DNA (right y-axis).

Extended Data Fig. 9 Effects of BE3 and SECURE BE3 variants on cell viability, structural model of rAPOBEC1 and extended sequence logos of off-target RNA edited sites.

a, Cell viability assay comparing HEK293T cells transfected with plasmid expressing nCas9–UGI–NLS, wild-type (WT) BE3, BE3-R33A, BE3-R33A/K34A or BE3-E63Q (n = 3 biologically independent samples per condition). Each dot represents one biological replicate (and is the mean of three technical replicates). All data points were normalized to the mean luminescence of the nCas9–UGI–NLS controls (set to 100%, grey dotted line) that were performed for each biological replicate experiment. The assay was performed on days 1, 2, 3 and 4 after plating (following sorting for all GFP-positive cells). Data shown as mean ± s.e.m. RLU, relative light unit; n.s., not significantly decreased compared to matched nCas9–UGI–NLS control; *P < 0.05, ***P < 0.001 for a significant decrease compared to matched nCas9–UGI–NLS control. Statistical significance was determined as described in Supplementary Methods. b, Structural model of rAPOBEC1 with locations of catalytic residues and the R33 and K34 positions that were altered in SECURE variants. A predicted rAPOBEC1 structure is shown that was generated with Protein Homology/analogY Recognition Engine v 2.0 (Phyre2)37 and visualized in PyMOL (v 1.8.2.1). The R33 and K34 residues mutated in the SECURE variants are shown in orange and blue, respectively. Catalytic site residues (H61, E63, C93 and C96) have previously been described19 and are shown in green. cf, Extended sequence logos for BE3- and ABEmax-induced RNA editing sites. Sequence logos derived with the nucleotides 100 base pairs upstream and downstream of the motifs edited in RNA by BE3 (ACW) or ABEmax (UA) are shown. Logos were derived from data for BE3 expression in HepG2 cells (c; Fig. 1c), BE3 expression in HEK293T cells (d; all GFP-sorted cells; Extended Data Fig. 2c), higher BE3 expression in HEK293T cells (e; top-5% GFP-sorted cells; Extended Data Fig. 4c), and ABEmax expression in HEK293T experiments (f; top 5% GFP-sorted cells; Fig. 3b). Analysis was done using RNA-seq data generated from cDNA; every T depicted should be considered a U in RNA.

Extended Data Table 1 Summary of RNA edits observed for all RNA-seq experiments

Supplementary information

Supplementary information

This file contains the Supplementary Methods, Supplementary Discussion, Supplementary References and a Supplementary Note which includes FACS raw data and gating examples for different experimental conditions.

Reporting Summary

Supplementary Tables

This file contains Supplementary Tables 1-36 and a Supplementary Table Guide.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grünewald, J., Zhou, R., Garcia, S.P. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437 (2019). https://doi.org/10.1038/s41586-019-1161-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1161-z

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research