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.

  • Letter
  • Published:

CRISPR DNA base editors with reduced RNA off-target and self-editing activities

Abstract

Cytosine or adenine base editors (CBEs or ABEs) can introduce specific DNA C-to-T or A-to-G alterations1,2,3,4. However, we recently demonstrated that they can also induce transcriptome-wide guide-RNA-independent editing of RNA bases5, and created selective curbing of unwanted RNA editing (SECURE)-BE3 variants that have reduced unwanted RNA-editing activity5. Here we describe structure-guided engineering of SECURE-ABE variants with reduced off-target RNA-editing activity and comparable on-target DNA-editing activity that are also among the smallest Streptococcus pyogenes Cas9 base editors described to date. We also tested CBEs with cytidine deaminases other than APOBEC1 and found that the human APOBEC3A-based CBE induces substantial editing of RNA bases, whereas an enhanced APOBEC3A-based CBE6, human activation-induced cytidine deaminase-based CBE7, and the Petromyzon marinus cytidine deaminase-based CBE Target-AID4 induce less editing of RNA. Finally, we found that CBEs and ABEs that exhibit RNA off-target editing activity can also self-edit their own transcripts, thereby leading to heterogeneity in base-editor coding sequences.

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

Access options

Buy this article

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

Fig. 1: Engineering of SECURE-ABE variants with reduced off-target RNA-editing activity.
Fig. 2: On-target DNA-editing activity of ABEmax, miniABEmax(K20A/R21A) and miniABEmax(V82G) in HEK293T cells.
Fig. 3: Transcriptome-wide off-target RNA-editing activity of CBEs with non-APOBEC1 cytidine deaminases in HEK293T cells.
Fig. 4: Self-editing generates heterogeneously edited CBE and ABE transcript sequences in HEK293T and HepG2 cells.

Similar content being viewed by others

Data availability

Plasmids encoding the SECURE-ABE and various CBE constructs shown in this work are available on Addgene under article number 28203996. The RNA-seq data used in this study have been deposited in the Gene Expression Omnibus (GEO) under accession GSE129894.

Targeted amplicon sequencing data have been deposited at the Sequence Read Archive BioProject accession number PRJNA553185. 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. 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  CAS  Google Scholar 

  3. 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  CAS  Google Scholar 

  4. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, aaf8729 (2016).

    Article  Google Scholar 

  5. Grünewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433–437 (2019).

    Article  Google Scholar 

  6. Gehrke, J. M. et al. An APOBEC3A–Cas9 base-editor with minimized bystander and off-target activities. Nat. Biotechnol. 36, 977–982 (2018).

    Article  CAS  Google Scholar 

  7. 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).

    Article  Google Scholar 

  8. 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 

  9. 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 

  10. 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 

  11. Losey, H. C., Ruthenburg, A. J. & Verdine, G. L. Crystal structure of Staphylococcus aureus tRNA adenosine deaminase TadA in complex with RNA. Nat. Struct. Mol. Biol. 13, 153–159 (2006).

    Article  CAS  Google Scholar 

  12. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR–Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).

    Article  CAS  Google Scholar 

  13. Wang, X. et al. Efficient base editing in methylated regions with a human APOBEC3A–Cas9 fusion. Nat. Biotechnol. 36, 946–949 (2018).

    Article  CAS  Google Scholar 

  14. Sharma, S., Patnaik, S. K., Kemer, Z. & Baysal, B. E. Transient overexpression of exogenous APOBEC3A causes C-to-U RNA editing of thousands of genes. RNA Biol. 14, 603–610 (2017).

    Article  Google Scholar 

  15. Fritz, E. L. et al. A comprehensive analysis of the effects of the deaminase AID on the transcriptome and methylome of activated B cells. Nat. Immunol. 14, 749–755 (2013).

    Article  CAS  Google Scholar 

  16. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275–278 (2019).

    Article  Google Scholar 

  17. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 5, eaax5717 (2019).

    Article  Google Scholar 

  18. Nishimasu, H. et al. Engineered CRISPR–Cas9 nuclease with expanded targeting space. Science 361, 1259–1262 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. 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 

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

    Article  CAS  Google Scholar 

  22. 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 

  23. 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 

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

    Article  CAS  Google Scholar 

Download references

Acknowledgements

J.K.J., J.G. and R.Z. are supported by the Defense Advanced Research Projects Agency (grant HR0011-17-2-0042). Support was also provided by the National Institutes of Health (grant RM1 HG009490 to J.K.J. and J.G., and grant R35 GM118158 to J.K.J. and M.J.A.). J.G. was supported by a research fellowship (GR 5129/1-1) of the German Research Foundation (DFG). J.K.J. is additionally supported by the Desmond and Ann Heathwood MGH Research Scholar Award. We thank G. Ciaramella for the suggestion to delete the wild-type TadA monomer from ABEmax. We thank A. Lapinaite for suggesting the overlay of E. coli and S. aureus TadA structures and S.J. Lee for technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

All wet lab experiments were performed by R.Z. and J.G. S.I., C.A.L., S.P.G. and M.J.A. performed computational analysis of the data. J.G. and J.K.J. conceived of and designed the study. J.G., M.J.A. and J.K.J. supervised the work. J.G. and J.K.J. wrote the initial manuscript draft 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, Excelsior Genomics, Pairwise Plants, Poseida Therapeutics, Transposagen Biopharmaceuticals and Verve Therapeutics (f/k/a Endcadia). The interests of J.K.J. were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. M.J.A. holds 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 and increase their precision.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 On-target DNA editing and additional data on the off-target RNA editing activities of ABEmax, miniABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G in HEK293T cells.

(a) Heat maps showing the on-target DNA editing efficiencies of nCas9 (Control), ABEmax, miniABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G each assessed with two gRNAs targeted to HEK site 2 and ABE site 16 and performed in triplicate. Note that these were performed with genomic DNA isolated from cells that were sorted for the RNA-seq experiments shown in Fig. 1c, d. Editing windows shown include only the most highly edited adenines and not the entire spacer sequence. Numbering at the bottom represents spacer position with 1 being the most PAM distal location. (b) Histograms showing the total number of RNA A-to-I edits observed (y-axis) with different editing efficiencies (x-axis) for ABEmax, miniABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G each tested with the HEK site 2, ABE site 16, and NT gRNAs. n = number of modified adenines. Experiments were performed in triplicate (data is derived from the same experiments as Fig. 1c, d). Dashed line = median; solid line = mean. Rep. = Replicate.

Supplementary Figure 2 Sequence logos for RNA adenines edited by ABEmax, miniABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G in HEK293T cells.

Sequence logos derived using all RNA-edited adenines (0–100]% or stratified RNA-edited adenines with high (80–100]%, middle (50–80]%, or low (0–50]% editing efficiencies induced by (a) ABEmax co-expressed with a HEK site 2, ABE site 16, or NT (non-targeting) gRNA or (b) miniABEmax co-expressed with a HEK site 2, ABE site 16, or NT gRNA or (c) miniABEmax-K20A/R21A or miniABEmax-V82G co-expressed with a HEK site 2, ABE site 16, or NT gRNA. Logos are shown for triplicate experiments from the same RNA-seq experiments displayed in Fig. 1c, d. n = total number of modified adenines. For strata that contained <50 edited adenines, we considered the motif analysis as not sufficiently powered and therefore presented these logos in a semi-transparent fashion or using all adenines (i.e., (0–100]% editing efficiency).

Supplementary Figure 3 Unexpected on-target editing of C bases by ABEs in HEK293T cells.

Heat maps showing (a) C-to-G (pink), (b) C-to-T (purple), and (c) C-to-A (green) DNA on-target editing efficiencies induced by nCas9 (Control), ABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G each co-expressed with HEK site 2, ABE site 7, or FANCF site 1 gRNAs (n=4 independent replicates). These were obtained from the same data presented in Fig. 2a, b but now including the entire spacer sequence and showing these other types of base edits. Numbering at the bottom represents spacer position with 1 being the most PAM-distal location.

Supplementary Figure 4 DNA off-target activities of ABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G in HEK293T cells.

Heat maps showing A-to-G DNA on-target (blue) and A-to-G DNA off-target (orange) editing efficiencies of nCas9 (Control), ABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G each co-expressed with HEK site 2, HEK site 3, or HEK site 4 gRNAs (n=4 independent replicates). Editing windows shown include the most highly edited adenines. Numbering at the bottom represents spacer position with 1 being the most PAM-distal location.

Supplementary Figure 5 Additional data showing transcriptome-wide off-target RNA editing activities and sequence logos of non-rAPOBEC1 CBEs in human HEK293T cells.

(a) Manhattan plots showing transcriptome-wide distribution of RNA edits induced by hA3A-BE3 (these are the same data shown as Jitter plots in Fig. 3b). Sequence logos derived using all RNA-edited cytosines (0–100]% or stratified RNA-edited cytosines with high (80–100]%, middle (50–80]%, or low (0–50]% editing efficiencies induced by hA3A-BE3 expressed with the RNF2 gRNA. n = total number of modified cytosines. (b) Histograms showing the total number of RNA C-to-U edits observed (y-axis) at different editing efficiencies (x-axis) with expression of hA3A-BE3, eA3A-BE3, hAID-BE3, or Target-AID co-expressed with the RNF2 gRNA. Data from triplicate experiments are shown (derived from the data shown as Jitter plots in Fig. 3b). Dashed line = median; solid line = mean. Rep. = Replicate. n = number of modified cytosines.

Supplementary Figure 6 Self-editing generates a diverse range of heterogeneously edited CBE and ABE transcript sequences in HEK293T and HepG2 cells.

(a) Plots showing C-to-U self-editing of the BE3-encoding RNA transcript observed with WT BE3 (with rAPOBEC1) expression in HEK293T cells (sorted for all GFP-positive cells) with two different gRNAs targeting sites in RNF2 and EMX1. Each dot represents an edited C and the color of the dot indicates the predicted type of mutation caused by a C-to-U edit at that position (Methods). n = total number of modified cytosines. The y-axis shows editing efficiencies for each C-to-U change and the x-axis represents the position of each C within the BE3 coding sequence (with the architecture of the editor shown schematically below but not displaying the NLS and linkers). Data were obtained by analyzing additional replicates from previously published RNA-seq experiments5. (b) Plots illustrating C-to-U self-editing observed with wild-type (WT) BE3 (with rAPOBEC1), SECURE-BE3 (R33A) and SECURE-BE3 (R33A/K34A) in HEK293T and HepG2 cells sorted for top 5% GFP signal with co-expression of the RNF2 gRNA. Data are shown as described in a. Data were obtained by analyzing additional replicates from previously published RNA-seq experiments5. (c) Plots depicting C-to-U self-editing observed in HEK293T cells expressing hA3A-BE3, eA3A-BE3, hAID-BE3, and Target-AID (sorted for top 5% GFP signal). Data are shown as described in a and were obtained using additional replicates from the RNA-seq experiments shown in Fig. 3b. (d) Plots showing A-to-I self-editing induced by expression of ABEmax, miniABEmax, miniABEmax-K20A/R21A, and miniABEmax-V82G (sorted for all GFP-positive cells) each with a gRNA targeting HEK site 2, ABE site 16, or a non-targeting gRNA (NT) in HEK293T cells. Data are shown as described in a and were obtained using additional replicates from the RNA-seq experiments shown in Fig. 1c, d. n = total number of modified adenines.

Supplementary Figure 7 Assessment of gRNA editing induced by CBEs and ABEs in human cells.

(a) Plots showing C-to-U edits on gRNA observed with WT BE3 expression in HEK293T cells (sorted for all GFP-positive cells) and gRNAs targeting sites in RNF2 or EMX1. Data were obtained by analyzing previously published RNA-seq experiments5. (b) Plots illustrating C-to-U edits on gRNA observed with wild-type (WT) BE3, SECURE-BE3 (R33A) and SECURE-BE3 (R33A/K34A) in HEK293T and HepG2 cells sorted for top 5% GFP signal with co-expression of the RNF2 gRNA. Data were obtained by analyzing previously published RNA-seq experiments5. (c) Plots depicting C-to-U edits on gRNA observed in HEK293T cells expressing hA3A-BE3, eA3A-BE3, hAID-BE3, or Target-AID (sorted for top 5% GFP signal) with the RNF2 gRNA. Data were obtained from the RNA-seq experiments shown in Fig. 3b. (d) Plots showing A-to-I edits on gRNA induced by expression of ABEmax, miniABEmax, miniABEmax-K20A/R21A, or miniABEmax-V82G (sorted for all GFP-positive cells) each assessed with gRNAs targeting HEK site 2, ABE site 16, or a non-targeting gRNA (NT) in HEK293T cells. Data were obtained from the RNA-seq experiments shown in Fig. 1c, d. For all plots, each dot represents an edited C or A. n = total number of modified Cs or As; the y-axis shows editing efficiencies for each C-to-U or A-to-I modifications and the x-axis represents the position of each C or A within the gRNA sequence (spacer and scaffold).

Supplementary information

Supplementary Figures

Supplementary Figs. 1–7, Supplementary Tables 1 and 2, and Supplementary Notes 1 and 2

Reporting Summary

Supplementary Table

Supplementary Table 3

Supplementary Table

Supplementary Table 4

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., Iyer, S. et al. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat Biotechnol 37, 1041–1048 (2019). https://doi.org/10.1038/s41587-019-0236-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0236-6

This article is cited by

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