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.

  • Review Article
  • Published:

Base editing: precision chemistry on the genome and transcriptome of living cells

A Correction to this article was published on 19 October 2018

This article has been updated

Abstract

RNA-guided programmable nucleases from CRISPR systems generate precise breaks in DNA or RNA at specified positions. In cells, this activity can lead to changes in DNA sequence or RNA transcript abundance. Base editing is a newer genome-editing approach that uses components from CRISPR systems together with other enzymes to directly install point mutations into cellular DNA or RNA without making double-stranded DNA breaks. DNA base editors comprise a catalytically disabled nuclease fused to a nucleobase deaminase enzyme and, in some cases, a DNA glycosylase inhibitor. RNA base editors achieve analogous changes using components that target RNA. Base editors directly convert one base or base pair into another, enabling the efficient installation of point mutations in non-dividing cells without generating excess undesired editing by-products. In this Review, we summarize base-editing strategies to generate specific and precise point mutations in genomic DNA and RNA, highlight recent developments that expand the scope, specificity, precision and in vivo delivery of base editors and discuss limitations and future directions of base editing for research and therapeutic applications.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Fig. 1: Distribution of human pathogenic genetic variants, including point mutations.
Fig. 2: Cytosine base editing.
Fig. 3: Adenine base editing in DNA and RNA.
Fig. 4: Overcoming targeting challenges associated with base editing.

Similar content being viewed by others

Change history

  • 19 October 2018

    The originally published article contained errors in reference numbering throughout table 1 (DNA base editors and their approximate editing windows) due to the unintended propagation of reference numbering from an earlier version of the table. The article has now been corrected online. The editors apologize for this error.

References

  1. Cohen, S. N., Chang, A. C., Boyer, H. W., Helling, R. B. Construction of biologically functional bacterial plasmids in vitro. Proc. Natl Acad. Sci. USA 70, 3240–3244 (1973).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012). This paper describes the isolation and purification of SpCas9 and the development of Cas9 nickases.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Jansen, R., Embden, J. D., Gaastra, W. & Schouls, L. M. Identification of genes that are associated with DNA repeats in prokaryotes. Mol. Microbiol. 43, 1565–1575 (2002).

    CAS  PubMed  Google Scholar 

  4. Garneau, J. E. et al. The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 468, 67–71 (2010).

    CAS  PubMed  Google Scholar 

  5. Cho, S. W., Kim, S., Kim, J. M. & Kim, J. S. Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat. Biotechnol. 31, 230–232 (2013).

    CAS  PubMed  Google Scholar 

  6. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Jinek, M. et al. RNA-programmed genome editing in human cells. eLife 2, e00471 (2013).

    PubMed  PubMed Central  Google Scholar 

  8. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-based technologies for the manipulation of eukaryotic genomes. Cell 168, 20–36 (2017).

    CAS  PubMed  Google Scholar 

  11. Doudna, J. A. & Charpentier, E. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    PubMed  Google Scholar 

  12. Sternberg, S. H. & Doudna, J. A. Expanding the biologist’s toolkit with CRISPR-Cas9. Mol. Cell 58, 568–574 (2015).

    CAS  PubMed  Google Scholar 

  13. Mali, P. et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol. 31, 833–838 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Bikard, D. et al. Programmable repression and activation of bacterial gene expression using an engineered CRISPR-Cas system. Nucleic Acids Res. 41, 7429–7437 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Cheng, A. W. et al. Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. 23, 1163–1171 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Gasiunas, G., Barrangou, R., Horvath, P. & Siksnys, V. Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl Acad. Sci. USA 109, E2579–E2586 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Jeggo, P. A. DNA breakage and repair. Adv. Genet. 38, 185–218 (1998).

    CAS  PubMed  Google Scholar 

  18. Rouet, P., Smih, F. & Jasin, M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol. Cell. Biol. 14, 8096–8106 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Lukacsovich, T., Yang, D. & Waldman, A. S. Repair of a specific double-strand break generated within a mammalian chromosome by yeast endonuclease I-SceI. Nucleic Acids Res. 22, 5649–5657 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Rudin, N., Sugarman, E. & Haber, J. E. Genetic and physical analysis of double-strand break repair and recombination in Saccharomyces cerevisiae. Genetics 122, 519–534 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Rouet, P., Smih, F. & Jasin, M. Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. Proc. Natl Acad. Sci. USA 91, 6064–6068 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Chapman, J. R., Taylor, M. R. & Boulton, S. J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497–510 (2012).

    CAS  PubMed  Google Scholar 

  23. Cox, D. B., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121–131 (2015). This review highlights the methods and challenges associated with therapeutic genome editing; it was published before the advent of base editing.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Paquet, D. et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125–129 (2016).

    CAS  PubMed  Google Scholar 

  25. Lin, S., Staahl, B. T., Alla, R. K. & Doudna, J. A. Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. eLife 3, e04766 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Goodwin, S., McPherson, J. D. & McCombie, W. R. Coming of age: ten years of next-generation sequencing technologies. Nat. Rev. Genet. 17, 333–351 (2016).

    CAS  PubMed  Google Scholar 

  27. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 38, 765–771 (2018).

    Google Scholar 

  28. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–D868 (2016).

    CAS  PubMed  Google Scholar 

  29. Landrum, M. J. et al. ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 42, D980–D985 (2014).

    CAS  PubMed  Google Scholar 

  30. Shin, H. Y. et al. CRISPR/Cas9 targeting events cause complex deletions and insertions at 17 sites in the mouse genome. Nat. Commun. 8, 15464 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. 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).

    CAS  PubMed  Google Scholar 

  32. Zhang, L. et al. Large genomic fragment deletions and insertions in mouse using CRISPR/Cas9. PLOS ONE 10, e0120396 (2015).

    PubMed  PubMed Central  Google Scholar 

  33. 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). This paper describes the development of the first DNA base editors, the cytosine base editors BE1, BE2 and BE3.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  Google Scholar 

  35. 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). This paper reports the evolution and engineering of the first DNA adenine base editor.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. 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). This paper describes improvements to our understanding of the mechanism of cytosine base editing in human cells and describes cytosine base editors with improved efficiency and product purity.

    PubMed  PubMed Central  Google Scholar 

  37. Nishimasu, H. et al. Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell 156, 935–949 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Li, X. et al. Base editing with a Cpf1-cytidine deaminase fusion. Nat. Biotechnol. 36, 324–327 (2018).

    CAS  PubMed  Google Scholar 

  39. Cox, D. B. T. et al. RNA editing with CRISPR-Cas13. Science 358, 1019–1027 (2017). This paper describes the development of the first Cas enzyme-guided RNA base editor, which can generate A-to-I mutations in RNA transcripts.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Montiel-Gonzalez, M. F., Vallecillo-Viejo, I., Yudowski, G. A. & Rosenthal, J. J. Correction of mutations within the cystic fibrosis transmembrane conductance regulator by site-directed RNA editing. Proc. Natl Acad. Sci. USA 110, 18285–18290 (2013). This paper describes a genetically encoded RNA base editor capable of generating A-to-I mutations in mRNA of mammalian cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Montiel-Gonzalez, M. F., Vallecillo-Viejo, I. C. & Rosenthal, J. J. An efficient system for selectively altering genetic information within mRNAs. Nucleic Acids Res. 44, e157 (2016).

    PubMed  PubMed Central  Google Scholar 

  42. Fukuda, M. et al. Construction of a guide-RNA for site-directed RNA mutagenesis utilising intracellular A-to-I RNA editing. Sci. Rep. 7, 41478 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Wettengel, J., Reautschnig, P., Geisler, S., Kahle, P. J. & Stafforst, T. Harnessing human ADAR2 for RNA repair — recoding a PINK1 mutation rescues mitophagy. Nucleic Acids Res. 45, 2797–2808 (2017).

    CAS  PubMed  Google Scholar 

  44. Vogel, P., Hanswillemenke, A. & Stafforst, T. Switching protein localization by site-directed RNA editing under control of light. ACS Synth. Biol. 6, 1642–1649 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Hanswillemenke, A., Kuzdere, T., Vogel, P., Jekely, G. & Stafforst, T. Site-directed RNA editing in vivo can be triggered by the light-driven assembly of an artificial riboprotein. J. Am. Chem. Soc. 137, 15875–15881 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Vogel, P. & Stafforst, T. Site-directed RNA editing with antagomir deaminases — a tool to study protein and RNA function. ChemMedChem 9, 2021–2025 (2014).

    CAS  PubMed  Google Scholar 

  47. Vogel, P., Schneider, M. F., Wettengel, J. & Stafforst, T. Improving site-directed RNA editing in vitro and in cell culture by chemical modification of the guideRNA. Angew. Chem. Int. Ed. 53, 6267–6271 (2014).

    CAS  Google Scholar 

  48. Stafforst, T. & Schneider, M. F. An RNA-deaminase conjugate selectively repairs point mutations. Angew. Chem. Int. Ed. 51, 11166–11169 (2012).

    CAS  Google Scholar 

  49. Vogel, P. et al. Efficient and precise editing of endogenous transcripts with SNAP-tagged ADARs. Nat. Methods 15, 535–538 (2018). In this paper, the authors address several of the limitations associated with RNA editing and generate a multiplexable, high-precision A-to-I RNA editor.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. 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).

    CAS  PubMed  Google Scholar 

  51. Kunz, C., Saito, Y. & Schar, P. DNA repair in mammalian cells — mismatched repair: variations on a theme. Cell. Mol. Life Sci. 66, 1021–1038 (2009).

    CAS  PubMed  Google Scholar 

  52. Pearl, L. H. Structure and function in the uracil-DNA glycosylase superfamily. Mutat. Res. 460, 165–181 (2000).

    CAS  PubMed  Google Scholar 

  53. Mol, C. D. et al. Crystal structure of human uracil-DNA glycosylase in complex with a protein inhibitor: protein mimicry of DNA. Cell 82, 701–708 (1995).

    CAS  PubMed  Google Scholar 

  54. Tang, W. & Liu, D. R. Rewritable multi-event analog recording in bacterial and mammalian cells. Science 360, eaap8992 (2018).

    PubMed  PubMed Central  Google Scholar 

  55. Krokan, H. E., Drablos, F. & Slupphaug, G. Uracil in DNA — occurrence, consequences and repair. Oncogene 21, 8935–8948 (2002).

    CAS  PubMed  Google Scholar 

  56. Lindahl, T. Instability and decay of the primary structure of DNA. Nature 362, 709–715 (1993).

    CAS  PubMed  Google Scholar 

  57. Yasui, M. et al. Miscoding properties of 2΄-deoxyinosine, a nitric oxide-derived DNA Adduct, during translesion synthesis catalyzed by human DNA polymerases. J. Mol. Biol. 377, 1015–1023 (2008).

    CAS  PubMed  Google Scholar 

  58. 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).

    CAS  PubMed  Google Scholar 

  59. Vogel, P. & Stafforst, T. Critical review on engineering deaminases for site-directed RNA editing. Curr. Opin. Biotechnol. 55, 74–80 (2018).

    PubMed  Google Scholar 

  60. Bass, B. L. & Weintraub, H. An unwinding activity that covalently modifies its double-stranded RNA substrate. Cell 55, 1089–1098 (1988).

    CAS  PubMed  Google Scholar 

  61. Matthews, M. M. et al. Structures of human ADAR2 bound to dsRNA reveal base-flipping mechanism and basis for site selectivity. Nat. Struct. Mol. Biol. 23, 426–433 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Yu, Y. T. et al. Internal modification of U2 small nuclear (sn)RNA occurs in nucleoli of Xenopus oocytes. J. Cell Biol. 152, 1279–1288 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Bass, B. L. RNA editing by adenosine deaminases that act on RNA. Annu. Rev. Biochem. 71, 817–846 (2002).

    CAS  PubMed  Google Scholar 

  64. Bazak, L. et al. A-To-I RNA editing occurs at over a hundred million genomic sites, located in a majority of human genes. Genome Res. 24, 365–376 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Keppler, A. et al. A general method for the covalent labeling of fusion proteins with small molecules in vivo. Nat. Biotechnol. 21, 86–89 (2003).

    CAS  PubMed  Google Scholar 

  66. Schneider, M. F., Wettengel, J., Hoffmann, P. C. & Stafforst, T. Optimal guideRNAs for re-directing deaminase activity of hADAR1 and hADAR2 in trans. Nucleic Acids Res. 42, e87 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Herbert, A. & Rich, A. The role of binding domains for dsRNA and Z-DNA in the in vivo editing of minimal substrates by ADAR1. Proc. Natl Acad. Sci. USA 98, 12132–12137 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Lehmann, K. A. & Bass, B. L. Double-stranded RNA adenosine deaminases ADAR1 and ADAR2 have overlapping specificities. Biochemistry 39, 12875–12884 (2000).

    CAS  PubMed  Google Scholar 

  69. Kuttan, A. & Bass, B. L. Mechanistic insights into editing-site specificity of ADARs. Proc. Natl Acad. Sci. USA 109, E3295–E3304 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Crick, F. H. Codon—anticodon pairing: the wobble hypothesis. J. Mol. Biol. 19, 548–555 (1966).

    CAS  PubMed  Google Scholar 

  71. Kume, H., Hino, K., Galipon, J. & Ui-Tei, K. A-To-I editing in the miRNA seed region regulates target mRNA selection and silencing efficiency. Nucleic Acids Res. 42, 10050–10060 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Seeburg, P. H. & Hartner, J. Regulation of ion channel/neurotransmitter receptor function by RNA editing. Curr. Opin. Neurobiol. 13, 279–283 (2003).

    CAS  PubMed  Google Scholar 

  73. Yang, J. H., Sklar, P., Axel, R. & Maniatis, T. Editing of glutamate receptor subunit B pre-mRNA in vitro by site-specific deamination of adenosine. Nature 374, 77–81 (1995).

    CAS  PubMed  Google Scholar 

  74. Nishikura, K. Functions and regulation of RNA editing by ADAR deaminases. Annu. Rev. Biochem. 79, 321–349 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Athanasiadis, A., Rich, A. & Maas, S. Widespread A-to-I RNA editing of Alu-containing mRNAs in the human transcriptome. PLOS Biol. 2, e391 (2004).

    PubMed  PubMed Central  Google Scholar 

  76. Hess, G. T. et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kim, K. et al. Highly efficient RNA-guided base editing in mouse embryos. Nat. Biotechnol. 35, 435–437 (2017).

    CAS  PubMed  Google Scholar 

  78. Ma, Y. et al. Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells. Nat. Methods 13, 1029–1035 (2016).

    CAS  PubMed  Google Scholar 

  79. Wang, L. et al. Enhanced base editing by co-expression of free uracil DNA glycosylase inhibitor. Cell Res. 27, 1289–1292 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Di Noia, J. & Neuberger, M. S. Altering the pathway of immunoglobulin hypermutation by inhibiting uracil-DNA glycosylase. Nature 419, 43–48 (2002).

    PubMed  Google Scholar 

  81. Radany, E. H. et al. Increased spontaneous mutation frequency in human cells expressing the phage PBS2-encoded inhibitor of uracil-DNA glycosylase. Mutat. Res. 461, 41–58 (2000).

    CAS  PubMed  Google Scholar 

  82. Hess, G. T., Tycko, J., Yao, D. & Bassik, M. C. Methods and applications of CRISPR-mediated base editing in eukaryotic genomes. Mol. Cell 68, 26–43 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Ryu, S. M. et al. Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nat. Biotechnol. 36, 536–539 (2018).

    CAS  PubMed  Google Scholar 

  84. Hua, K., Tao, X., Yuan, F., Wang, D. & Zhu, J. K. Precise A·T to G·C base editing in the rice genome. Mol. Plant 11, 627–630 (2018).

    CAS  PubMed  Google Scholar 

  85. Yan, F. et al. Highly efficient A·T to G·C base editing by Cas9n-guided tRNA adenosine deaminase in rice. Mol. Plant 11, 631–634 (2018).

    CAS  PubMed  Google Scholar 

  86. Hu, J. H. et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 556, 57–63 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Lau, A. Y., Wyatt, M. D., Glassner, B. J., Samson, L. D. & Ellenberger, T. Molecular basis for discriminating between normal and damaged bases by the human alkyladenine glycosylase, AAG. Proc. Natl Acad. Sci. USA 97, 13573–13578 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Kouzminova, E. A. & Kuzminov, A. Patterns of chromosomal fragmentation due to uracil-DNA incorporation reveal a novel mechanism of replication-dependent double-stranded breaks. Mol. Microbiol. 68, 202–215 (2008).

    CAS  PubMed  Google Scholar 

  89. Liu, Z. et al. Highly efficient RNA-guided base editing in rabbit. Nat. Commun. 9, 2717 (2018).

    PubMed  PubMed Central  Google Scholar 

  90. Yeh, W. H., Chiang, H., Rees, H. A., Edge, A. S. B. & Liu, D. R. In vivo base editing of post-mitotic sensory cells. Nat. Commun. 9, 2184 (2018).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Kang, B. C. et al. Precision genome engineering through adenine base editing in plants. Nat. Plants 4, 427–431 (2018).

    CAS  PubMed  Google Scholar 

  93. Rees, H. A. et al. Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat. Commun. 8, 15790 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  95. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490–495 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Lee, J. K. et al. Directed evolution of CRISPR-Cas9 to increase its specificity. Nat. Commun. 9, 3048 (2018).

    PubMed  PubMed Central  Google Scholar 

  97. Yamanaka, S. et al. Apolipoprotein B mRNA-editing protein induces hepatocellular carcinoma and dysplasia in transgenic animals. Proc. Natl Acad. Sci. USA 92, 8483–8487 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Okazaki, I. M. et al. Constitutive expression of AID leads to tumorigenesis. J. Exp. Med. 197, 1173–1181 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Burns, M. B. et al. APOBEC3B is an enzymatic source of mutation in breast cancer. Nature 494, 366–370 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Liu, Z. et al. Efficient generation of mouse models of human diseases via ABE- and BE-mediated base editing. Nat. Commun. 9, 2338 (2018).

    PubMed  PubMed Central  Google Scholar 

  101. Gehrke, J. M. et al. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. https://doi.org/10.1038/nbt.4199 (2018).

  102. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–191 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Kim, E. et al. In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat. Commun. 8, 14500 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Muller, M. et al. Streptococcus thermophilus CRISPR-Cas9 systems enable specific editing of the human genome. Mol. Ther. 24, 636–644 (2016).

    PubMed  PubMed Central  Google Scholar 

  106. Lee, C. M., Cradick, T. J. & Bao, G. The Neisseria meningitidis CRISPR-Cas9 system enables specific genome editing in mammalian cells. Mol. Ther. 24, 645–654 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Kleinstiver, B. P. et al. Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol. 33, 1293–1298 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Kleinstiver, B. P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    PubMed  PubMed Central  Google Scholar 

  109. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371–376 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Jiang, W. et al. BE-PLUS: a new base editing tool with broadened editing window and enhanced fidelity. Cell Res. 28, 855–861 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Kim, S., Bae, T., Hwang, J. & Kim, J. S. Rescue of high-specificity Cas9 variants using sgRNAs with matched 5’ nucleotides. Genome Biol. 18, 218 (2017).

    PubMed  PubMed Central  Google Scholar 

  113. Nishimasu, H. et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science https://doi.org/10.1126/science.aas9129 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Hua, K., Tao, X. & Zhu, J. K. Expanding the base editing scope in rice by using Cas9 variants. Plant Biotechnol. J. https://doi.org/10.1111/pbi.12993 (2018).

  115. Yang, L. et al. Increasing targeting scope of adenosine base editors in mouse and rat embryos through fusion of TadA deaminase with Cas9 variants. Protein Cell 9, 814–819 (2018).

    PubMed  PubMed Central  Google Scholar 

  116. Wang, X. et al. Efficient base editing in methylated regions with a human APOBEC3A-Cas9 fusion. Nat. Biotechnol. https://doi.org/10.1038/nbt.4198 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Pinello, L. et al. Analyzing CRISPR genome-editing experiments with CRISPResso. Nat. Biotechnol. 34, 695–697 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Clement, K. et al. Analysis and comparison of genome editing using CRISPResso2. Preprint at https://www.biorxiv.org/content/early/2018/08/15/392217 (2018).

  119. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144–149 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Wheeler, L. C., Lim, S. A., Marqusee, S. & Harms, M. J. The thermostability and specificity of ancient proteins. Curr. Opin. Struct. Biol. 38, 37–43 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Zafra, M. P. et al. Optimized base editors enable efficient editing in cells, organoids and mice. Nat. Biotechnol. 36, (888–893 (2018).

    Google Scholar 

  122. Midoux, P., Pichon, C., Yaouanc, J. J. & Jaffres, P. A. Chemical vectors for gene delivery: a current review on polymers, peptides and lipids containing histidine or imidazole as nucleic acids carriers. Br. J. Pharmacol. 157, 166–178 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Bodles-Brakhop, A. M., Heller, R. & Draghia-Akli, R. Electroporation for the delivery of DNA-based vaccines and immunotherapeutics: current clinical developments. Mol. Ther. 17, 585–592 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Thomas, C. E., Ehrhardt, A. & Kay, M. A. Progress and problems with the use of viral vectors for gene therapy. Nat. Rev. Genet. 4, 346–358 (2003).

    CAS  PubMed  Google Scholar 

  125. Zuris, J. A. et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol. 33, 73–80 (2015).

    CAS  PubMed  Google Scholar 

  126. Wang, M. et al. Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc. Natl Acad. Sci. USA 113, 2868–2873 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Kim, S., Kim, D., Cho, S. W., Kim, J. & Kim, J. S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012–1019 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Miao, C. H. et al. Nonrandom transduction of recombinant adeno-associated virus vectors in mouse hepatocytes in vivo: cell cycling does not influence hepatocyte transduction. J. Virol. 74, 3793–3803 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Dong, J. Y., Fan, P. D. & Frizzell, R. A. Quantitative analysis of the packaging capacity of recombinant adeno-associated virus. Hum. Gene Ther. 7, 2101–2112 (1996).

    CAS  PubMed  Google Scholar 

  130. Lai, Y. et al. Efficient in vivo gene expression by trans-splicing adeno-associated viral vectors. Nat. Biotechnol. 23, 1435–1439 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 553, 217–221 (2018).

    CAS  PubMed  Google Scholar 

  132. Park, D. S. et al. Targeted base editing via RNA-guided cytidine deaminases in Xenopus laevis embryos. Mol. Cells 40, 823–827 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Liang, P. et al. Effective gene editing by high-fidelity base editor 2 in mouse zygotes. Protein Cell 8, 601–611 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Li, G. et al. Highly efficient and precise base editing in discarded human tripronuclear embryos. Protein Cell 8, 776–779 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Liang, P. et al. Correction of beta-thalassemia mutant by base editor in human embryos. Protein Cell 8, 811–822 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Ma, Y. et al. Highly efficient and precise base editing by engineered dCas9-guide tRNA adenosine deaminase in rats. Cell Discov. 4, 39 (2018).

    PubMed  PubMed Central  Google Scholar 

  137. Zhang, Y. et al. Programmable base editing of zebrafish genome using a modified CRISPR-Cas9 system. Nat. Commun. 8, 118 (2017).

    PubMed  PubMed Central  Google Scholar 

  138. Tanaka, S. et al. In vivo targeted single-nucleotide editing in zebrafish. Sci. Rep. 8, 11423 (2018).

    PubMed  PubMed Central  Google Scholar 

  139. Schenk, B. et al. MPDU1 mutations underlie a novel human congenital disorder of glycosylation, designated type If. J. Clin. Invest. 108, 1687–1695 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Zeng, Y. et al. Correction of the Marfan syndrome pathogenic FBN1 mutation by base editing in human cells and heterozygous embryos. Mol. Ther. https://doi.org/10.1016/j.ymthe.2018.08.007 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Chadwick, A. C., Wang, X. & Musunuru, K. In vivo base editing of PCSK9 (proprotein convertase subtilisin/kexin type 9) as a therapeutic alternative to genome editing. Arterioscler. Thromb. Vasc. Biol. 37, 1741–1747 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Roccio, M., Hahnewald, S., Perny, M. & Senn, P. Cell cycle reactivation of cochlear progenitor cells in neonatal FUCCI mice by a GSK3 small molecule inhibitor. Sci. Rep. 5, 17886 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Kuscu, C. et al. CRISPR-STOP: gene silencing through base-editing-induced nonsense mutations. Nat. Methods 14, 710–712 (2017).

    CAS  PubMed  Google Scholar 

  145. Billon, P. et al. CRISPR-mediated base editing enables efficient disruption of eukaryotic genes through induction of STOP codons. Mol. Cell 67, 1068–1079 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927–930 (2018).

    CAS  PubMed  Google Scholar 

  147. Ihry, R. J. et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939–946 (2018).

    CAS  PubMed  Google Scholar 

  148. Aguirre, A. J. et al. Genomic copy number dictates a gene-independent cell response to CRISPR/Cas9 targeting. Cancer Discov. 6, 914–929 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Roukos, V. & Misteli, T. The biogenesis of chromosome translocations. Nat. Cell Biol. 16, 293–300 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Gapinske, M. et al. CRISPR-SKIP: programmable gene splicing with single base editors. Genome Biol. 19, 107 (2018).

    PubMed  PubMed Central  Google Scholar 

  151. Hur, J. K. et al. Targeted mutagenesis in mice by electroporation of Cpf1 ribonucleoproteins. Nat. Biotechnol. 34, 807–808 (2016).

    CAS  PubMed  Google Scholar 

  152. Sung, Y. H. et al. Knockout mice created by TALEN-mediated gene targeting. Nat. Biotechnol. 31, 23–24 (2013).

    CAS  PubMed  Google Scholar 

  153. Sung, Y. H. et al. Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Res. 24, 125–131 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Farzadfard, F. & Lu, T. K. Emerging applications for DNA writers and molecular recorders. Science 361, 870–875 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Tang, W., Hu, J. H. & Liu, D. R. Aptazyme-embedded guide RNAs enable ligand-responsive genome editing and transcriptional activation. Nat. Commun. 8, 15939 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Farzadfard, F. et al. Single-nucleotide-resolution computing and memory in living cells. Preprint at https://www.biorxiv.org/content/early/2018/02/15/263657 (2018).

  157. Yin, K., Gao, C. & Qiu, J. L. Progress and prospects in plant genome editing. Nat. Plants 3, 17107 (2017).

    CAS  PubMed  Google Scholar 

  158. Voytas, D. F. & Gao, C. Precision genome engineering and agriculture: opportunities and regulatory challenges. PLOS Biol. 12, e1001877 (2014).

    PubMed  PubMed Central  Google Scholar 

  159. Svitashev, S. et al. Targeted mutagenesis, precise gene editing, and site-specific gene insertion in maize using Cas9 and guide RNA. Plant Physiol. 169, 931–945 (2015).

    PubMed  PubMed Central  Google Scholar 

  160. Endo, M., Mikami, M. & Toki, S. Biallelic gene targeting in rice. Plant Physiol. 170, 667–677 (2016).

    CAS  PubMed  Google Scholar 

  161. Zong, Y. et al. Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat. Biotechnol. 35, 438–440 (2017).

    CAS  PubMed  Google Scholar 

  162. Shimatani, Z. et al. Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat. Biotechnol. 35, 441–443 (2017).

    CAS  PubMed  Google Scholar 

  163. Li, C. et al. Expanded base editing in rice and wheat using a Cas9-adenosine deaminase fusion. Genome Biol. 19, 59 (2018).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.R.L. acknowledges support from Defense Advanced Research Projects Agency (DARPA) HR0011-17-2-0049; the Ono Pharma Foundation; US National Institutes of Health (NIH) RM1 HG009490, R01 EB022376, U01 AI142756 and R35 GM118062; and Howard Hughes Medical Institute (HHMI). H.A.R. is supported by the Kilpatrick Educational fund from the Chemistry and Chemical Biology Department, Harvard University. The authors thank J.K. Joung, F. Zhang, A. Raguram, W.-H. Yeh, T. Huang, K. Zhao and W. Tang for their helpful comments.

Author information

Authors and Affiliations

Authors

Contributions

Both authors contributed to all aspects of the manuscript.

Corresponding author

Correspondence to David R. Liu.

Ethics declarations

Competing interests

D.R.L. is a consultant and co-founder of Beam Therapeutics, Editas Medicine and Pairwise Plants, which are all companies that use genome-editing technologies. H.A.R. declares no competing financial interests. Both authors have no competing non-financial interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Glossary

Guide RNA

Short RNA sequence comprising a scaffold for binding to the necessary CRISPR-associated (Cas) enzyme and a variable spacer region that defines the target site for the enzyme. In natural CRISPR systems, the guide RNA is often made of two molecules of RNA with complementarity. Engineered ‘single-guide’ RNAs connecting the two natural guide RNA components are often accepted by Cas enzymes.

Protospacer adjacent motif

(PAM). A small region of nucleotides in the target DNA sequence adjacent to the sequence specified by a guide RNA. The PAM is not specified in the guide RNA, but CRISPR-associated (Cas) enzymes do not bind or cleave a sequence unless they are next to the appropriate PAM.

Cas9 nickase

A catalytically disabled mutant of a Cas9 enzyme that is able to create a single-stranded DNA break but not a double-stranded DNA break.

Activity window

The region of DNA or RNA, typically defined by the number of nucleotides from the protospacer adjacent motif (PAM), in which a particular base editor acts to induce efficient point mutations. The activity window for most base editors is approximately four to five nucleotides wide.

Protospacer

A region in a guide RNA of 15–25 nucleotides in length that specifies the target RNA or DNA locus.

Proximal off-target editing

Unwanted editing of bases that occurs outside of the activity window but is found nearby (for example, 100 nucleotides upstream or downstream of) the target site.

Distal off-target editing

Unwanted editing of bases residing in locations of the genome or transcriptome unrelated to (for example, >100 nucleotides away from) the target site of the base editor.

Cas13b

A class 2, type VI RNA-guided RNase from the CRISPR system. Variants from several species have been characterized. It catalyses site-specific cleavage of single-stranded RNA.

Wobble position

The third nucleotide in a codon.

Base-editing product purity

The term used to describe the spectrum of mutations induced by a particular base-editing technology. Low product purity occurs when a target base is mutated to bases other than the desired point mutation or when small insertions or deletions are generated in addition to the desired edit; for example, C-to-G or C-to-A edits, rather than the desired C-to-T edit, from a cytosine base editor.

SpCas9

An RNA-guided endonuclease variant isolated from the CRISPR system of Streptococcus pyogenes. It catalyses site-specific cleavage of double-stranded DNA at sites with an NGG protospacer adjacent motif (PAM).

SaCas9

An RNA-guided endonuclease variant isolated from the CRISPR system of Staphylococcus aureus. It catalyses site-specific cleavage of double-stranded DNA at sites with an NNGRRT protospacer adjacent motif (PAM).

Bystander editing

Editing of a non-target base that resides in the activity window of a particular base editor and guide RNA. Bystander editing occurs in addition to editing of the target base.

Cas12a

A class 2, type V RNA-guided endonuclease from the CRISPR system. Variants from several species have been characterized. It catalyses site-specific cleavage of double-stranded DNA at sites with a TTTV protospacer adjacent motif (where V is A, C or G).

Mosaicism

A state in which two or more cell populations with distinct genotypes are present in the same organism and derived from a single fertilized egg.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

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). https://doi.org/10.1038/s41576-018-0059-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-018-0059-1

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